Language selection

Search

Patent 3148275 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3148275
(54) English Title: THERAPY GUIDANCE AND/OR THERAPY MONITORING FOR TREATMENT OF SHOCK
(54) French Title: ORIENTATION DE THERAPIE ET/OU SURVEILLANCE DE THERAPIE PERMETTANT LE TRAITEMENT D'UN CHOC
Status: Examination Requested
Bibliographic Data
(51) International Patent Classification (IPC):
  • G01N 33/48 (2006.01)
  • A61P 9/00 (2006.01)
  • G01N 33/53 (2006.01)
  • A61K 39/395 (2006.01)
(72) Inventors :
  • BERGMANN, ANDREAS (Germany)
(73) Owners :
  • 4TEEN4 PHARMACEUTICALS GMBH (Germany)
(71) Applicants :
  • 4TEEN4 PHARMACEUTICALS GMBH (Germany)
(74) Agent: MARKS & CLERK
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2020-08-28
(87) Open to Public Inspection: 2021-03-04
Examination requested: 2022-09-08
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/EP2020/074134
(87) International Publication Number: WO2021/038078
(85) National Entry: 2022-02-16

(30) Application Priority Data:
Application No. Country/Territory Date
19194729.0 European Patent Office (EPO) 2019-08-30
19201098.1 European Patent Office (EPO) 2019-10-02

Abstracts

English Abstract

Subject matter of the present invention is a method for predicting or diagnosing a refractory shock in a subject that either runs into shock or that has developed shock, wherein said method is comprising the steps: * determining the level of DPP3 in a sample of bodily fluid of said subject; * comparing said level of determined DPP3 to a predetermined threshold, wherein said subject is predicted to run into refractory shock or is diagnosed as having refractory shock if said determined level of DPP3 is above said predetermined threshold. Further subject matter relates to vasopressors, angiotensin-receptor agonists and/or precursors thereof, inhibitors of the activity of DPP3 and anti-ADM antibodies for use in therapy of shock in a subject that either runs into shock or that has developed shock.


French Abstract

La présente invention a pour objet un procédé de prédiction ou de diagnostic d'un choc réfractaire chez un sujet qui soit entre en état de choc soit a développé un choc, ledit procédé comprenant les étapes consistant : * à déterminer le niveau de DPP3 dans un échantillon de fluide corporel dudit sujet; * à comparer ledit niveau de DPP3 déterminé à un seuil prédéterminé, il est prédit que ledit sujet subira un choc réfractaire ou est diagnostiqué comme présentant un choc réfractaire si ledit niveau déterminé de DPP3 se situe au-dessus dudit seuil prédéterminé. L'invention concerne en outre des agents vasopresseurs, des agonistes du récepteur de l'angiotensine et/ou des précurseurs de ces derniers, des inhibiteurs de l'activité de la DPP3 et des anticorps anti-ADM destinés à être utilisés dans la thérapie du choc chez un sujet qui soit entre en état de choc soit a développé un choc.

Claims

Note: Claims are shown in the official language in which they were submitted.


WO 2021/038078 100
PCT/EP2020/074134
CLAIMS
1. A method for predicting or diagnosing a refractory shock in a subject that
either mns into shock or
that has developed shock, wherein said method is comprising the steps:
= determining the level of DPP3 in a sample of bodily fluid of said
subject;
= comparing said level of determined DPP3 to a predetermined threshold,
wherein said subject is predicted to run into refractory shock or is diagnosed
as having refractory
shock if said determined level of DPP3 is above said predetermined threshold.
io 2. A method for predicting or diagnosing a refractory shock in a subject
that either runs into shock or
that has developed shock according to claim 1, wherein said shock is selected
from the group
comprising shock due to hypovolemia, cardiogenic shock, obstructive shock and
distributive shock,
in particular cardiogenic shock or septic shock.
is 3. A method for predicting or diagnosing a refractory shock in a subject
that either runs into shock or
that has developed shock according to dim 1 or 2, wherein said shock is a
vasopressor-resistant
shock.
4. A method for predicting or diagnosing a refractory shock in a subject that
either rims into shock or
20 that has developed shock according to claims 1 to 3, wherein
= in case of cardiogenic shock said subject may have suffered an acute
coronary syndrome (e.g.
acute myocardial infarction) or wherein said subject has heart failure (e.g.
acute decompensated
heart failure), myocarditis, arrhythmia, cardiomyopathy, valvular heart
disease, aortic dissection
with acute aortic stenosis, traumatic chordal rupture or massive puhnonary
embolism, or
25 = in case of hypovolemic shock said subject may have suffered a
hemorrhagic disease including
gastrointestinal bleed, trauma, vascular etiologies (e.g. ruptured abdominal
aortic aneurysm,
tumor eroding into a major blood vessel) and spontaneous bleeding in the
setting of
anticoagulant use or a non-hemorrhagic disease including vomiting, diarrhea,
renal loss, skin
losses/insensible losses (e.g. burns, heat stroke) or third-space loss in the
setting of pancreatitis,
30 cirrhosis, intestinal obstruction, trauma, or
= in case of obstructive shock said patient may have suffered a cardiac
tamponade, tension
pneumothorax, pulmonary embolism or aortic stenosis, or
= in case of distributive shock said patient may have septic shock,
neumgenic shock, anaphylactic
shock or shock due to adrenal crisis.
CA 03148275 2022-2-16

WO 2021/038078 101
PCT/EP2020/074134
5. A method for predicting or diagnosing a refiactory shock in a subject that
either runs into shock or
that has developed shock according to claims 1 to 4, wherein said method is
used for initiation
and/or termination and/ or stratification and/or guidance of treatment.
6. A method for predicting or diagnosing a refractory shock in a subject that
either runs into shock or
that has developed shock according to any of claims 1 to 5, wherein a
treatment is initiated and/or
maintained and/or withheld and/ or terminated if said determined level of DPP3
is above said
predetermined threshold.
io
7. A method for predicting or
diagnosing a refractory shock in a subject that either runs into shock or
that has developed shock according to claim 6, wherein said treatment is
selected from the group of
vasopressors, Angiotensin-Receptor-Agonists and/or precursors thereof,
inhibitors of the DPP3
activity and anti-adrenomedullin antibodies or anti-adrenomedullin antibody
fragments.
8. A method for predicting or diagnosing a refractory shock in a subject that
either runs into shock or
that has developed shock according to any of claims 1 to 7, wherein either the
level of DPP3
pmtein and/or the level of active DPP3 is determined and compared to a
predetermined threshold,
wherein the level of DPP3 is determined by contacting said sample of bodily
fluid with a capture
binder that binds specifically to DPP3.
9. A method for predicting or diagnosing a refractory shock in a subject that
either runs into shock or
that has developed shock according to any of claims 1 to 8, wherein a
treaiment with Angiotensin-
Receptor-Agonists and/ or precursors thereof and/or inhibitors of the DPP3
activity is initiated
and/or continued when the level of DPP3 in said sample is above a certain
threshold and/or
wherein a treatment with vasopressors is withheld and/ or terminated if said
determined level of
DPP3 is above said predetermined threshold.
10. A method for predicting or diagnosing a refractory shock in a subject that
either runs into shock or
that has developed shock according to any of claims 1 to 9, wherein a
treatment with vasopressors
is initiated and/or continued when the level of DPP3 in said sample is below a
certain threshold
and/or wherein a treatment with Angiotensin-Receptor-Agonists and/ or
precursors thereof and/or
inhibitors of the DPP3 activity is withheld and/ or terminated if the said
determined level of DPP3
is below said predetermined threshold.
CA 03148275 2022-2-16

WO 2021/038078 102
PCT/EP2020/074134
11.A method for predicting or diagnosing a refractory shock in a subject that
either runs into shock or
that has developed shock according to claim 9, wherein in addition the level
of Pro-adrenomedullin
or fragments thereof is determined and wherein treatment with an anti-ADM
antibody or anti-ADM
antibody fragment is initiated and/or continued when the level of Pro-
adrenomedullin or fragments
thereof in said sample is above a certain threshold and/or wherein a treatment
with an anti-ADM
antibody or anti-ADM antibody fragment is withheld and/ or terminated if the
said determined
level of Pm-adrenomedullin or fragments thereof is below said predetermined
threshold.
12.A method for predicting or diagnosing a refractory shock in a subject that
either runs into shock or
io that has developed shock according to claim 9, wherein treatment with
an anti-ADM antibody or
anti-ADM antibody fragment and/or Angiotensin-Receptor-Agonists and/ or
precursors thereof
and/or inhibitors of the DPP3 activity is initiated and/or continued if the
level of Pro-
adrenomedullin or fragments thereof in said sample is above a certain
threshold and said
determined level of DPP3 is above said predetermined threshold of DPP3.
13Nasopressor for use in therapy of shock in a subject that either runs into
shock or that has
developed shock, wherein said subject has a level of DPP3 in a sample of
bodily fluid of said
subject that is below a predetermined threshold, when determined by a method
according to any of
claims 1 ¨ 11.
14.Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either runs into shock
or that has developed shock, wherein said subject has a level of DPP3 in a
sample of bodily fluid of
said subject that is above a predetermined threshold when determined by a
method according to
any of claims 1 ¨ 11, wherein the inhibitor of the activity of DPP3 is
selected from the group
comprising anti-DPP3 antibody or anti-DPP3 antibody fragment or anti-DPP3 non-
Ig scaffold.
15,Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either runs into shock
or that has developed shock according to claim 14, wherein said inhibitor is
administered in
combination with an Angiotensin-Receptor-Agonist and/or precursor thereof.
16.Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either runs into shock
or that has developed shock according to claim 15, wherein said Angiotensin-
Receptor-Agonist
and/ or precursor thereof is selected from the group comprising angiotensin I,
angiotensin II,
angiotensin III, angiotensin IV, in particular angiotensin H.
CA 03148275 2022-2-16

WO 2021/038078 103
PCT/EP2020/074134
ILA method of treatment of shock in a subject that either runs into shock or
that has developed shock,
the method comprising adrninistering vasopressor to said subject, wherein said
subject has a level
of DPP3 in a sample of bodily fluid of said subject that is below a
predetermined threshold, when
determined by a method according to any of claims 1 ¨ 11.
18.Method of treatment of shock in a subject that either mns into shock or
that has developed shock,
the method comprising administering inhibitor of DPP3 activity to said
subject, wherein said
subject has a level of DPP3 in a sample of bodily fluid of said subject that
is above a predetermined
threshold when determined by a method according to any of claims I ¨ 11.
ufr
19.Method of treatment of shock in a subject that either rims into shock or
that has developed shock
according to claim 18, the method comprising administering an inhibitor of
DPP3 activity to said
subject, wherein said inhibitor is administered in combination with an
Angiotensin-Receptor-
Agonist and/ or a precursor thereof
20.Method of treatment of shock in a subject that either runs into shock or
that has developed shock
according to claims 17-18, the method comprising administering Angiotensin-
Receptor-Agonists
and/ or precursors thereof and/or inhibitor of DPP3 activity to said subject,
wherein the treatment
with said Angiotensin-Receptor-Agonists and/or inhibitors of the DPP3 activity
is initiated and/or
continued when the level of DPP3 in a sample of said subject is above a
certain threshold and/or
wherein a treatment with vasopressors is withheld and/ or terminated if said
determined level of
DPP3 is above said predetermined threshold.
21.Method of treatment of shock in a subject that either runs into shock or
that has developed shock
according to claims 17-18, the method comprising administering vasopressor to
said subject,
wherein the treatment with said vasopressors is initiated and/or continued
when the level of DPP3
in a sample of bodily fluid of said subject is below a certain threshold
and/or wherein a treatment
with Angiotensin-Receptor-Agonists and/ or precursors thereof and/or
inhibitors of the DPP3
activity is withheld and/ or terminated if the said determined level of DPP3
is below said
predetermined threshold.
22.Method of treatment of shock in a subject that either runs into shock or
that has developed shock
according to claim 19, the method comprising administering Angiotensin-
Receptor-Agonists and/
or precursors thereof and/or inhibitor of DPP3 activity to said subject, and
wherein in addition the
level of Pro-adrenomedullin or fragments thereof is determined and wherein
treatment with said
CA 03148275 2022-2-16

WO 2021/038078 104
PCT/EP2020/074134
anti-ADM antibody or anti-ADM antibody fragment is initiated and/or continued
when the level of
Pro-adrenomedullin or fragments thereof in said sample is above a certain
threshold and/or wherein
a treatment with an anti-ADM antibody or anti-ADM antibody fragment is
withheld and/ or
terminated if the said determined level of Pro-adrenomedullin or fragments
thereof is below said
predetermined threshold.
23.Method of treatment of shock in a subject that either mns into shock or
that has developed shock
according to claim 20, the method comprising administering vasopressor to said
subject, and
wherein in addition the level of Pro-adrenomedullin or fragments thereof is
determined and
io wherein treatment with said anti-ADM antibody or anti-ADM antibody
fragment is initiated and/or
continued when the level of Pro-adrenomedullin or fragments thereof in said
sample is above a
certain threshold and/or wherein a treannent with an anti-ADM antibody or anti-
ADM antibody
fragment is withheld and/ or terminated if the said determined level of Pro-
adrenomedullin or
fragments thereof is below said predetermined threshold.
24.Method of treatment of shock in a subject that either runs into shock or
that has developed shock
according to claim 21 and 22, the method comprising administering anti-ADM
antibody or anti-
ADM antibody fragment to said subject, wherein in addition the level of Pro-
adrenomedullin or
fragments thereof is determined and wherein treatment with said anti-ADM
antibody or anti-ADM
antibody fragment is initiated and/or continued when the level of Pro-
adrenomedullin or fragments
thereof in said sample is above a certain threshold and/or wherein a treatment
with an anti-ADM
antibody or anti-ADM antibody fragment is withheld and/ or terminated if the
said determined
level of Pm-adrenomedullin or fragments thereof is below said predetermined
threshold.
25.A method for prognosing an outcome and/or the risk of an adverse event in a
subject that has
developed refractory shock, wherein said method is comprising the steps:
= determining the level of DPP3 in a sample of bodily fluid of said
subject;
= comparing said level of determined DPP3 to a predetermined threshold,
= correlating said level of DPP3 with said risk of an adverse event in said
subject, wherein an
elevated level above a certain threshold is predictive for an enhanced risk of
said adverse events
or,
= correlating said level of DPP3 with success of a therapy or intervention
in said subject wherein
a level below a certain threshold is predictive for a success of therapy or
intervention.
CA 03148275 2022-2-16

Description

Note: Descriptions are shown in the official language in which they were submitted.


WO 2021/038078 1
PCT/EP2020/074134
Therapy Guidance and/or Therapy Monitoring for treatment of shock
The present invention is directed to a method for predicting or diagnosing a
refractory shock
in a subject that either runs into shock or that has developed shock, and
vasopressors,
angiotensin-receptor agonists and/or precursors thereof, inhibitors of DPP3
and anti-ADM
antibodies for use in therapy of shock in a subject that either runs into
shock or that has
developed shock, and methods of treating said shock or refractory shock.
STATE OF THE ART
Dipeptidyl peptidase 3 ¨ also known as Dipeptidyl aminopeptidase III,
Dipeptidyl
io arylamidase III, Dipeptidyl peptidase III, Enkephalinase B or red cell
angiotensinase; short
name: DPP3, DPPIII ¨ is a metallopeptidase that removes dipeptides from
physiologically
active peptides, such as enkephalins and angiotensins.
DPP3 was first identified and its activity measured in extracts of purified
bovine anterior
pituitary by Ellis & Nuenke 1967. The enzyme, which is listed as EC 3.4.14.4,
has a
IS molecular mass of about 83 kDa and is highly conserved in procaiyotes
and eucaryotes
(Prajapati & Chauhan 2011). The amino acid sequence of the human variant is
depicted in
SEQ ID NO 1. Dipeptidyl peptidase III is a mainly cytosolic peptidase which is
ubiquitously
expressed. Despite lacking a signal sequence, a few studies reported
membranous activity
(Lee &Snyder 1982).
20 DPP3 is a zinc-depending exo-peptidase belonging to the peptidase family
M49. It has a
broad substrate specificity for oligopeptides from three/ four to ten amino
acids of various
compositions and is also capable of cleaving after proline. DPP3 is known to
hydrolyze
dipeptides from the N-terminus of its substrates, including angiotensin II,
Ill and IV; Leu- and
Met-enkephalin; endomorphin 1 and 2. The metallopeptidase DPP3 has its
activity optimum
25 at pH 8.0-9.0 and can be activated by addition of divalent metal ions,
such as Co2F and Mg2F.
Structural analysis of DPP3 revealed the catalytic motifs HELLGH (hDPP3 450-
455) and
EECRAE (hDPP3 507-512), as well as following amino acids, that are important
for substrate
binding and hydrolysis: Glu316, Tyr, 318, Asp366, Asn391, Asn394, His568,
Arg572,
Arg577, Lys666 and Arg669 (Prajapati & Chauhan 2011; Kumar et aL 2016;
numbering
30 refers to the sequence of human DPP3, see SEQ ID NO. 1). Considering all
known amino
acids or sequence regions that are involved in substrate binding and
hydrolysis, the active site
of human DPP3 can be defined as the area between amino acids 316 and 669.
CA 03148275 2022-2-16

WO 2021/038078 2
PCT/EP2020/074134
The most prominent substrate of DPP3 is angiotensin II (Ang II), the main
effector of the
renin¨angiotensin system (RAS). The RAS is activated in cardiovascular
diseases (Dostal et
al. 1997. J Mot Cell Cardio/;29:2893-902; Roks et at 1997. Heart Vessels.
Suppl 12:119-
24), sepsis, and septic shock (Correa et at 2015. Crit Care 2015:19:98). Ang
II, in particular,
has been shown to modulate many cardiovascular functions including the control
of blood
pressure and cardiac remodeling.
The exact biological function of DPP3 in cellular physiology is not
understood, but recent
findings indicate its role not only in in protein metabolism but also in pain
modulation and
inflammatory processes (Prajapati & Chauhan 2011). In addition, DPP3 reduced
blood
io pressure in Ang,Thinfused hypertensive mice, without changing heart rate
(Pang et al. 2016).
However normotensive mice had no alteration in blood pressure, when DPP3 was
injected.
Blood pressure reduction by administration of DPP3 together with an
angiotensin-receptor
antagonist in Ang II¨infused mice was similar to that of DPP3 injection alone
or angiotensin-
receptor antagonist alone (Pang et at 2016. Hypertension 68:630-41).
Angiotensin II (AngII) is a peptide hormone naturally produced by the body
that regulates
blood pressure via vasoconstriction and sodium reabsorption. The hemodynamic
effects of
Ang II administration have been the subject of numerous clinical studies,
demonstrating
significant effects on systemic and renal blood flow (Harrison-Bernard 2009.
Adv. Physiol
Ed,,. 33(4): 270),
Ang11-treatment is currently discussed for its beneficial effect in
vasodilatory shock and septic
shock (Khanna. A. et at, 2017; Antonucci, E. et aL 2017, Tumlin, IA. et at
2018). Patients
with vasodilatory shock (80% in septic shock) treated with angiotensin II were
more likely to
survive to 28 days and showed a significant correction of hypotension (Khanna,
A. et at,
2017; Turn/in, IA. et at 2018).
It is assumed that the Angiotensin-converting enzyme (ACE) is highly
dysregulated in shock
patients leading to an altered angiotensin I/II ratio) and that angiotensin II
infusion may
compensate for such dysregulation (Tutnlin. J.A. et at 2018).
Recently, two assays were generated, characterized, and validated to
specifically detect DPP3
in human bodily fluids (e.g., blood, plasma, serum): a luminescence
immunoassay (LIA) to
detect DPP3 protein concentration and an enzyme capture activity assay (ECA)
to detect
specific DPP3 activity (Rehfeld et aL 2019 JALAI 3(6):943-953). A washing step
in both
methods removes all interfering substances before the actual detection of DPP3
protein or
CA 03148275 2022-2-16

WO 2021/038078 3
PCT/EP2020/074134
activity is performed. Both methods are highly specific and allow the
reproducible detection
of DPP3 in blood samples.
The object of the present invention is the provision of a method for
predicting or diagnosing a
refractory shock in a subject that either runs into shock or that has
developed shock.
Another object of the invention is a provision of Vasopressors, angiotensin-
receptor agonists
and/or precursors thereof, inhibitors of DPP3 and anti-ADM antibodies for use
in therapy of
shock in a subject that either runs into shock or that has developed shock.
Subject of the present invention is a method for predicting or diagnosing a
refractory shock in
a subject that either runs into shock or that has developed shock, wherein
said method is
io comprising the steps:
= determining the level of DPP3 in a sample of bodily fluid of said
subject;
= comparing said level of determined DPP3 to a predetermined threshold,
wherein said subject is predicted to run into refractory shock or is diagnosed
as having
refractory shock if said determined level of DPP3 is above said predetermined
threshold.
Subject of the present invention is a method for predicting or diagnosing a
refractory shock in
a subject that either runs into shock or that has developed shock, wherein
said method is
comprising the steps:
= determining the level of DPP3 in a sample of bodily fluid of said
subject, and
= comparing said level of determined DPP3 to a predetermined threshold, and
= determining the level of pro-adrenomedullin of fragments thereof in a
sample of
bodily fluid of said subject, and
= comparing said level of determined pro-adrenomedullin or fragments
thereof to a
predetermined threshold,
wherein said subject is predicted to run into refractory shock or is diagnosed
as having
refractory shock if said determined level of pro-adrenomedullin or fragments
thereof and/or
said level of DPP3 is above said predetermined threshold.
In a specific embodiment of said method for predicting or diagnosing a
refractory shock in a
subject that either runs into shock or that has developed shock, said shock is
selected from the
CA 03148275 2022-2-16

WO 2021/038078 4
PCT/EP2020/074134
group comprising shock due to hypovolemia, cardiogenic shock, obstructive
shock and
distributive shock.
In another specific embodiment of said method for predicting or diagnosing a
refractory
shock in a subject that either runs into shock or that has developed shock,
said shock is
selected from the group comprising shock due to hypovolemia, cardiogenic
shock, obstructive
shock and distributive shock, in particular cardiogenic or septic shock.
In a specific embodiment of said method for predicting or diagnosing a
refractory shock in a
ro
subject that either runs into shock or
that has developed shock, said shock is selected from the
group comprising:
= in case of cardiogenic shock said subject has suffered an acute coronary
syndrome
(e.g. acute myocardial infarction) or has heart failure (e.g. acute
decompensated heart
failure), myocarditis, arrhythmia, cardiomypathy, valcular heart disease,
aortic
dissection with acute aortic stenosis, traumatic chordal rupture Of massive
pulmonary
embolism, or
= in case of hypovolemic shock said subject may have suffered a hemorrhagic
disease
including gastrointestinal bleed, trauma, vascular etiologies (e.g. ruptured
abdominal
aortic aneurysm, tumor eroding into a major blood vessel) and spontaneous
bleeding
in the setting of anticoagulant use or a non-hemorrhagic disease including
vomiting,
diarrhea, renal loss, skin losses/insensible losses (e.g. bums, heat stroke)
or third-space
loss in the setting of pancreatitis, cirrhosis, intestinal obstruction,
trauma, or
= in case of obstructive shock said patient may have suffered a cardiac
tamponade,
tension pneumothorax, pulmonary embolism or aortic stenosis, or
= in case of distributive shock said patient has septic shock, neurogenic
shock,
anaphylactic shock or shock due to adrenal crisis.
In another embodiment of said method for predicting or diagnosing a refractory
shock in a
subject that either runs into shock or that has developed shock, said method
is used for
initiation and/or termination and/ or stratification and/or guidance of
treatment.
CA 03148275 2022-2-16

WO 2021/038078 5
PCT/EP2020/074134
As used herein, the term "therapy guidance" or "guidance of treatment" refers
to application
of certain therapies or medical interventions based on the value of one or
more biomarkers
and/or clinical parameter and/or clinical scores.
The term "therapy monitoring" in the context of the present invention refers
to the monitoring
and/or adjustment of a therapeutic treatment of said patient, for example by
obtaining
feedback on the efficacy of the therapy.
The term "therapy stratification" in particular relates to grouping or
classifying patients into
io different groups, such as therapy groups that receive or do not receive
therapeutic measures
depending on their classification.
The term "prediction" means correlating a probability of an outcome with a
result obtained in
the measurement of an analyte. An example of this is the measurement of a
certain marker,
such as DPP3, in a sample, the measured level of which is correlated with the
probability of a
subject that either runs into shock or that has developed shock, to develop a
refractory shock.
The present invention also comprises a method for short-term prediction of a
refractory shock
in a subject that either runs into shock or that has developed shock, whereby
short-term means
a period of equal or less than 14 days, preferably equal or less than 7 days,
more preferred
equal or less than 3 days, most preferred equal or less than 48 hours.
In another embodiment of said method for predicting or diagnosing a refractory
shock in a
subject that either runs into shock or that has developed shock, said
treatment is initiated
and/or maintained and/or withheld and/or terminated if said determined level
of DPP3 is
above said predetermined threshold.
In another embodiment of said method for predicting or diagnosing a refractory
shock in a
subject that either runs into shock or that has developed shock, said
treatment is initiated
and/or maintained and/or withheld and/or terminated if said determined level
of DPP3 and/ or
pro-adrenomedullin and/ or fragments thereof is above said predetermined
threshold.
In a preferred embodiment of said method for predicting or diagnosing a
refractory shock in a
subject that either runs into shock or that has developed shock, said
treatment is selected from
CA 03148275 2022-2-16

WO 2021/038078 6
PCT/EP2020/074134
the group of vasopressors, angiotensin receptor agonists or precursors
thereof, and/or
inhibitors of the DPP3 activity.
In another preferred embodiment of said method for predicting or diagnosing a
refractory
shock in a subject that either runs into shock or that has developed shock,
said treatment is
selected from the group of vasopressors, Angiotensin-Receptor-Agonists and/or
precursors
thereof, inhibitors of the DPP3 activity and anti-adrenomedullin antibodies or
anti-
adrenomedullin antibody fragments.
io In another embodiment of said method for predicting or diagnosing a
refractory shock in a
subject that either runs into shock or that has developed shock either the
level of DPP3 protein
and/or the level of active DPP3 is determined and compared to a predetermined
threshold.
In another embodiment of said method for predicting or diagnosing a refractory
shock in a
subject that either runs into shock or that has developed shock the level of
DPP3 is
determined by contacting said sample of bodily fluid with a capture binder
that binds
specifically to DPP3.
In another preferred embodiment of said method for predicting or diagnosing a
refractory
shock in a subject that either runs into shock or that has developed shock,
said capture binder
for determining the level of DPP3 may be selected from the group of antibody,
antibody
fragment or non-IgG scaffold.
In a specific embodiment of said method method for predicting or diagnosing a
refractory
shock in a subject that either runs into shock or that has developed shock,
said capture binder
is an antibody.
In another specific embodiment of said method for predicting or diagnosing a
refractory
shock in a subject that either runs into shock or that has developed shock,
said capture binder
is a monoclonal antibody.
In another embodiment of said method for predicting or diagnosing a refractory
shock in a
subject that either runs into shock or that has developed shock, said sample
of bodily fluid is
selected from the group of whole blood, plasma, and serum.
CA 03148275 2022-2-16

WO 2021/038078 7
PCT/EP2020/074134
In a specific embodiment of said method for predicting or diagnosing a
refractory shock in a
subject that either runs into shock or that has developed shock, said method
of diagnosing or
predicting is conducted at least twice.
Shock is characterized by decreased oxygen delivery and/or increased oxygen
consumption or
inadequate oxygen utilization leading to cellular and tissue hypoxia. It is a
life-threatening
condition of circulatory failure and most commonly manifested as hypotension
(systolic blood
pressure less than 90 mm Hg or MAP less than 65 mmHg). Shock is divided into
four main
io types based on the underlying cause: hypovolemic, cardiogenic,
obstructive, and distributive
shock (Vincent and De Backer 2014. N. Engl. J. Med. 370(6): 583).
Hypovolemic shock is characterized by decreased intravascular volume and can
be divided
into two broad subtypes: hemorrhagic and nonhemorrhagic. Common causes of
hemorrhagic
hypovolemic shock include gastrointestinal bleed, trauma, vascular etiologies
(e.g. ruptured
abdominal aortic aneurysm, tumor eroding into a major blood vessel) and
spontaneous
bleeding in the setting of anticoagulant use. Common causes of nonhemorrhagic
hypovolemic
shock include vomiting, diarrhea, renal loss, skin losses/insensible losses
(e.g. burns, heat
stroke) or third-space loss in the setting of pancreatitis, cirrhosis,
intestinal obstruction,
trauma. For review see Koya and Paul 2018. Shock StatPearls [Internet].
Treasure Island
(FL): StatPearls Publishing; 2019-2018 Oct 27.
Cardiogenic shock (CS) is defined as a state of critical endorgan
hypoperfusion due to
reduced cardiac output. Notably, CS forms a spectrum that ranges from mild
hypoperfusion to
profound shock. Established criteria for the diagnosis of CS are: (i) systolic
blood pressure,
<90 mmHg for >30 min or vasopressors required to achieve a blood pressure >90
mmHg; (ii)
pulmonary congestion or elevated left-ventricular filling pressures; (iii)
signs of impaired
organ perfusion with at least one of the following criteria: (a) altered
mental status; (b) cold,
clammy skin; (c) oliguria (< 0.5 mL/kg/h or <30 mL/h), (d) increased serum-
lactate (Reynolds
and Hochman 2008. Circulation 117: 686-697). Acute myocardial infarction (AMI)
with
subsequent ventricular dysfunction is the most frequent cause of CS accounting
for
approximately 80% of cases. Mechanical complications such as ventricular
septal (4%) or free
wall rupture (2%), and acute severe mitral regurgitation (7%) are less
frequent causes of CS
after AMI. (Hochman et al. 2000. J Am Coll Ccrrdiol 36: 1063-1070). Non-AMI-
related CS
may be caused by decompensated valvular heart disease, acute myocarditis,
arrhythmias, etc.
with heterogeneous treatment options. This translates in 40 000 to 50 000
patients per year in
CA 03148275 2022-2-16

WO 2021/038078 8
PCT/EP2020/074134
the USA and 60 000 to 70 000 in Europe. Despite advances in treatment mainly
by early
revascularization with subsequent mortality reduction, CS remains the leading
cause of death
in AMI with mortality rates still approaching 40-50% according to recent
registries and
randomized trials (Goldberg et al. 2009. Circulation 119: 1211-1219).
Obstructive shock is due to a physical obstruction of the great vessels or the
heart itself
Several conditions can result in this form of shock (e.g. cardiac tamponade,
tension
pneumothorax, pulmonary embolism, aortic stenosis). For review see Koya and
Paul 2018.
Shock StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2019-
2018 Oct 27.
According to the cause, there are four types of distributive shock: neurogenic
shock
io (decreased sympathetic stimulation leading to decreased vasal tone),
anaphylactic shock,
septic shock and shock due to adrenal crisis. In addition to sepsis,
distributive shock can be
caused by systemic inflammatory response syndrome (SIRS) due to conditions
other than
infection such as pancreatitis, bums or trauma. Other causes include, toxic
shock syndrome
(TSS), anaphylaxis (a sudden, severe allergic reaction), adrenal insufficiency
(acute
worsening of chronic adrenal insufficiency, destruction or removal of the
adrenal glands,
suppression of adrenal gland function due to exogenous steroids,
hypopituitarism and
metabolic failure of hormone production), reactions to drugs or toxins, heavy
metal
poisoning, hepatic (liver) insufficiency and damage to the central nervous
system. For review
see Koya and Paul 2018. Shock Ski/Pearls [Internet]. Treasure Island (FL):
StatPearls
Publishing; 2019-2018 Oct 27.
In general, refractory shock has been defined as requirement of noradrenaline
infusion of >0.5
lig/kg/min despite adequate volume resuscitation. Mortality in these patients
may be as high
as 94% and the assessment and management of these patients requires a much
more
aggressive approach for survival. The term õrefractory shock" is used when the
tissue
perfusion cannot be restored with the initial corrective measures employed
(e.g. vasopressors)
and may therefore be referred to as õhigh vasopressor-dependent" or
õvasopressor-resistant"
shock (Udupa and Shetty 2018. Indian J Respir Care 7: 6711172). Patients with
refractory
shock may have features of inadequate perfusion such as hypotension (mean
arterial blood
pressure <65 mmHg), tachycardia, cold peripheries, prolonged capillary refill
time, and
tachypnea consequent to the hypoxia and acidosis. Fever may be seen in septic
shock. Other
signs of hypoperfusion such as altered sensorium, hyperlactatemia, and
oliguria may also be
seen. These well Llknown signs of shock are not helpful in identifying whether
the problem is
at the pump (heart) or circuitry (vessels and tissues). Different types of
shock can coexist, and
CA 03148275 2022-2-16

WO 2021/038078 9
PCT/EP2020/074134
all forms of shock can become refractory, as evidenced by unresponsiveness to
highll dose
vasopressors (Udzipa and Sheri)) 2018. Indian f Respir Care 7: 67072).
In a specific embodiment of the invention said refractory shock is vasopressor-
resistant, that
is defined by unresponsiveness of the patient to high dose vasopressors (e.g.
>0.5 Fig/kg/min
noradrenaline).
The consequence of the diagnosis or the prediction that a subject either runs
into a
vasopressor-resistant refractory shock or is diagnosed with a vasopressor-
resistant refractory
io shock maybe the administration of treatment like administration of
angiotensin II inhibitors
and/or DPP3 inhibitors. Furthermore, another consequence maybe the withholding
of
vasopressors.
In a preferred embodiment of said method for predicting a refractory shock, in
particular a
vasopressor-resitant shock, in a subject that potentially runs into shock,
wherein it is predicted
whether said subject runs into a refractory shock, in particular a vasopressor-
resitant shock,
from the point of time the sample is taken within 14 days, or in particular
within 10 days, or
in particular within 7 days, or in particular within 5 days, or in particular
within 48 hours, or
in particular within 24 hours, or in particular between 24 and 48 hours.
Another embodiment of the present invention relates to a method for predicting
or diagnosing
a refractory shock in a subject that either runs into shock or that has
developed shock, wherein
a treatment with Angiotensin-Receptor-Agonists and/or precursors thereof
and/or inhibitors of
the DPP3 activity is initiated and/or continued when the level of DPP3 in said
sample is above
a certain threshold and/or wherein a treatment with vasopressors is withheld
and/ or
terminated if said determined level of DPP3 is above said predetermined
threshold.
Another specific embodiment of the present invention relates to a method for
predicting or
diagnosing a refractory shock in a subject that either runs into shock or that
has developed
shock, wherein a treatment with vasopressors is initiated and/or continued
when the level of
DPP3 in said sample is below a certain threshold and/or wherein a treatment
with
Angiotensin-Receptor-Agonists and/or precursors thereof and/or inhibitors of
the DPP3
activity is withheld and/ or terminated if the said determined level of DPP3
is below said
predetermined threshold.
CA 03148275 2022-2-16

WO 2021/038078 10
PCT/EP2020/074134
A further embodiment of the present invention relates to a method for
predicting or
diagnosing a refractory shock in a subject that either runs into shock or that
has developed
shock, wherein a treatment with vasopressors is initiated and/or continued
when the level of
DPP3 in said sample is below a certain threshold and/or wherein a treatment
with
Angiotensin-Receptor-Agonists and/or precursors thereof and/or inhibitors of
the DPP3
activity is withheld and/ or terminated if the said determined level of DPP3
is below said
predetermined threshold and the level of Pro-adrenomedullin or fragments
thereof is
determined and wherein treatment with an anti-ADM antibody or anti-ADM
antibody
fragment is initiated and/or continued when the level of Pro-adrenomedullin or
fragments
thereof in said sample is above a certain threshold and/or wherein a treatment
with an anti-
ADM antibody or anti-ADM antibody fragment is withheld and/ or terminated if
the said
determined level of Pro-adrenomedullin or fragments thereof is below said
predetermined
threshold.
A further embodiment of the present invention relates to a method for
predicting or
diagnosing a refractory shock in a subject that either runs into shock or that
has developed
shock according to the present invention, wherein
= said subject may have suffered an acute coronary syndrome (e.g. acute
myocardial
infarction) or wherein said subject has heart failure (e_g_ acute
decompensated heart
failure), myocarditis, arrhythmia, cardiomyopathy, valvular heart disease,
aortic
dissection with acute aortic stenosis, traumatic chordal rupture or massive
pulmonary
embolism (in case of cardiogenic shock), or
= said subject may have suffered a hemorrhagic disease including
gastrointestinal bleed,
trauma, vascular etiologies (e.g. ruptured abdominal aortic aneurysm, tumor
eroding
into a major blood vessel) and spontaneous bleeding in the setting of
anticoagulant use
or a non-hemorrhagic disease including vomiting, diarrhea, renal loss, skin
losses/insensible losses (e.g. bums, heat stroke) or third-space loss in the
setting of
pancreatitis, cirrhosis, intestinal obstruction, trauma, (in case of
hypovolemic shock)
or
= said patient may have suffered a cardiac tamponade, tension pneumothorax,

pulmonary embolism or aortic stenosis (in case of obstructive shock), or
= said patient may have septic shock, neurogenic shock, anaphylactic shock
or shock
due to adrenal crisis (in case of distributive shock.)
CA 03148275 2022-2-16

WO 2021/038078 11
PCT/EP2020/074134
A further embodiment of the present invention relates to a method for
predicting or
diagnosing a refractory shock in a subject that either runs into shock or that
has developed
shock according to the present invention, wherein said method is used for
initiation and/or
termination and/ or stratification and/or guidance of treatment
A further embodiment of the present invention relates to a method for
predicting or
diagnosing a refractory shock in a subject that either runs into shock or that
has developed
shock according to the present invention, wherein a treatment is initiated
and/or maintained
io and/or withheld and/ or terminated if said determined level of DPP3 is
above said
predetermined threshold.
Subject matter of the present invention are Vasopressors, Angiotensin-Receptor-
Agonists
and/or precursors thereof, inhibitors of the DPP3 activity and/or anti-
adrenomedullin
antibodies or anti-adrenomedullin antibody fragments for use in a method of
prevention or
treatment of a refractory shock in a subject that either either runs into
shock or that has
developed shock, wherein said prediction and/or diagnosis of said refractory
shock has been
determined by a method for predicting or diagnosing a refractory shock in a
subject that either
runs into shock or that has developed shock according to the present
invention. In one
embodiment of the beforementioned methods either the level of DPP3 protein
and/or the level
of active DPP3 is determined and compared to a predetermined threshold,
wherein the level
of DPP3 is determined by contacting said sample of bodily fluid with a capture
binder that
binds specifically to DPP3.
Subject matter of the present invention are Angiotensin-Receptor-Agonists
and/or precursors
thereof, inhibitors of the DPP3 activity for use in a method of prevention or
treatment of a
refractory shock in a subject that either either runs into shock or that has
developed shock,
wherein said prediction and/or diagnosis of said refractory shock has been
determined by a
method for predicting or diagnosing a refractory shock in a subject that
either runs into shock
or that has developed shock according to the present invention, wherein a
treatment with
Angiotensin-Receptor-Agonists and/ or precursors thereof and/or inhibitors of
the DPP3
activity is initiated and/or continued when the level of DPP3 in said sample
is above a certain
threshold and/or wherein a treatment with vasopressors is withheld and/ or
terminated if said
determined level of DPP3 is above said predetermined threshold.
CA 03148275 2022-2-16

WO 2021/038078 12
PCT/EP2020/074134
Subject matter of the present invention are Angiotensin-Receptor-Agonists
and/or precursors
thereof, inhibitors of the DPP3 activity for use in a method of prevention or
treatment of a
refractory shock in a subject that either either runs into shock or that has
developed shock,
wherein said prediction and/or diagnosis of said refractory shock has been
determined by a
method for predicting or diagnosing a refractory shock in a subject that
either runs into shock
or that has developed shock according to the present invention, wherein a
treatment with
vasopressors is initiated and/or continued when the level of DPP3 in said
sample is below a
certain threshold and/or wherein a treatment with Angiotensin-Receptor-
Agonists and/ or
io precursors thereof and/or inhibitors of the DPP3 activity is withheld
and/ or terminated if the
said determined level of DPP3 is below said predetermined threshold.
Subject matter of the present invention is an anti-ADM antibody or anti-ADM
antibody
fragment for use in a method of prevention or treatment of a refractory shock
in a subject that
either either runs into shock or that has developed shock, wherein said
prediction and/or
diagnosis of said refractory shock has been determined by a method for
predicting or
diagnosing a refractory shock in a subject that either runs into shock or that
has developed
shock according to the present invention, wherein in addition the level of Pro-
adrenomedullin
or fragments thereof is determined and wherein treatment with an anti-ADM
antibody or anti-
ADM antibody fragment is initiated and/or continued when the level of Pro-
adrenomedullin
or fragments thereof in said sample is above a certain threshold and/or
wherein a treatment
with an anti-ADM antibody or anti-ADM antibody fragment is withheld and/ or
terminated if
the said determined level of Pro-adrenomedullin or fragments thereof is below
said
predetermined threshold.
Another embodiment of the present invention relates to a method for predicting
or diagnosing
a refractory shock in a subject that either runs into shock or that has
developed shock, wherein
treatment with an anti-ADM antibody or anti-ADM antibody fragment and/or
Angiotensin-
Receptor-Agonists and/or precursors thereof and/or inhibitors of the DPP3
activity is initiated
and/or continued if the level of Pro-adrenomedullin or fragments thereof in
said sample is
above a certain threshold and said determined level of DPP3 is above said
predetermined
threshold of DPP3,
CA 03148275 2022-2-16

WO 2021/038078 13
PCT/EP2020/074134
Subject matter of the present invention is an anti-ADM antibody or anti-ADM
antibody
fragment and/or an inhibitor of the DPP3 activity for use in a method of
prevention or
treatment of a refractory shock in a subject that either either runs into
shock or that has
developed shock, wherein said prediction and/or diagnosis of said refractory
shock has been
determined by a method for predicting or diagnosing a refractory shock in a
subject that either
runs into shock or that has developed shock according to the present
invention, wherein
treatment with an anti-ADM antibody or anti-ADM antibody fragment and/or
Angiotensin-
Receptor-Agonists and/ or precursors thereof and/or inhibitors of the DPP3
activity is initiated
and/or continued if the level of Pro-adrenomedullin or fragments thereof in
said sample is
io above a certain threshold and said determined level of DPP3 is above
said predetermined
threshold of DPP3.
In a specific embodiment of the invention the threshold is within a threshold
range for plasma
DPP3 that is between 20 and 200 ng/ml, preferably between 25 and 150 ng/ml,
even more
preferred between 30 and 100 ng/ml, even more preferred between 35 and 75
ng/ml, most
preferred a threshold of 50 ng/mL is applied.
In particular the refractory shock of said patient is a vasopressor-resistant
shock.
The peptide adrenomedullin (ADM) was described as a novel hypotensive peptide
comprising
52 amino acids, which had been isolated from a human pheochromocytoma
(Kitamura et teL
1993. Biochemical and Biophysical Research Communications 192 (2,): 553-560).
Cleavage
of the N-terminal signal sequence of 21 amino acids from pre-pro-
Adrenomedullin (pre-
proADM) results in the precursor peptide pro-Adrenomedullin (proADM) (SEQ ID
No.: 31).
Pro-ADM is further cleaved into PAMP (SEQ ID No. 32), MR-proADM (SEQ ID No.
33),
ADM-Gly (SEQ ID No.: 35) and CT-proADM (SEQ ID No. 36). The mature
adrenomedullin
peptide is an amidated peptide (ADM-NH2), which comprises 52 amino acids (SEQ
ID No:
34) and which comprises the amino acids 95 to 146 of pre-proADM, from which it
is formed
by proteolytic cleavage. Mature ADM, bio-ADM and ADM-NH2 is used synonymously
throughout this application and is a molecule according to SEQ ID No.: 34.
In one embodiment of the subject matter of the present invention said fragment
of
pro-Adrenomedullin is selected from a group comprising PAMP (SEQ ID No. 32),
MR-proADM (SEQ ID No. 33), amidated ADM (SEQ ID No. 34), ADM-Gly (SEQ ID No.:
35) and CT-proADM (SEQ ID No. 36).
CA 03148275 2022-2-16

WO 2021/038078 14
PCT/EP2020/074134
ADM may be regarded as a polyfunctional regulatory peptide It is released into
the
circulation partially in an inactive form extended by glycine (Kitamura et at
1998. Bloc/win.
Biophys. Res. Commum 244(2): 551-555). There is also a binding protein (Pio et
at 2001.
The Journal of Biological Chemistry 276(15): 12292-12300), which is specific
for ADM and
probably likewise modulates the effect of ADM.
Those physiological effects of ADM as well as of PAMP, which are of primary
importance in
the investigations to date, were the effects influencing blood pressure. Thus,
ADM is an
effective vasodilator. It was found that the concentrations of ADM, which can
be measured in
io the circulation and other biological fluids, are in a number of
pathological states, significantly
above the concentrations to be found in healthy control persons. Thus, the ADM
level in
patients with congestive heart failure, myocardial infarction, kidney
diseases, hypertensive
disorders, diabetes mellitus, in the acute phase of shock and in sepsis and
septic shock are
significantly increased, although to different extents. The PAMP
concentrations are also
increased in some of said pathological states, but the plasma levels are
reduced relative to
ADM (Eto et at 2001. Peptides 22: 1693-1711).
Furthermore, it is known that unusual high concentrations of ADM are to be
observed in
sepsis or in septic shock (Eto et at 2001. Peptides 22: 1693-1711; Hirata et
at 1996. Journal
of Clinical Endocrinology and Metabolism 81(4): 1449-1453; Ehlenz et al. 1997.
Eycp Clin
Endocrinol Diabetes 105: 156-162; Tomoda et at 2001. Peptides 22: 1783-1794;
Ueda et al
1999 Am. .1. Respir. Cr/i. Care Med.160: 132-136; Wang et at 2001. Peptides
22: 1835-
1840). The findings are related to the typical hemodynamic changes which are
known as
typical phenomena of the course of a disease in patients with sepsis and other
severe
syndromes, such as, for example, SIRS. Adrenomedullin plays pivotal roles
during sepsis
development (Wang. Shock 1998. 10(5):383-384; Wang et at 1998. Archives of
surgery
133(12): 1298-1304) and in numerous acute and chronic diseases (Parlapiano et
at 1999.
European Review for Medical and Pharmacological Sciences 3:53-61; Hinson et at
2000
Endocrine Reviews 21(2):138-167).
MR-proADM has been identified as a prognostic marker that is able to stratify
the mortality
risk in sepsis patients with different degrees of organ failure. It may be
helpful in the early
identification and individual risk assessment of sepsis and may also
facilitate the subsequent
clinical management of sepsis and septic shock (for review see Nal et at 2018.
Healthcare
6: 110).
CA 03148275 2022-2-16

WO 2021/038078 15
PCT/EP2020/074134
Several methods were described to measure circulating levels of ADM: either
ADM directly
or indirectly by determining a more stable fragment of its cognate precursor
peptide. Recently
a method was published, describing an assay to measure circulating mature ADM
(Weber et
aL 2017. JALM 2: 222-233).
Other methods to quantify fragments derived from the ADM precursor have been
described,
e.g. the measurement of MR-proADM (Morgenthaler et al. 2005 Clin Chem
51(J0,):1823-9),
PAMP (Washimine et al. 1994. Bloc/tern Biophys Res Commun 202(2):1081-7) and
CT-
io proADM (EP 2 111 552). A commercial homogeneous time-resolved
fluoroimmunoassay for
the measurement of MR-proADM in plasma on a fully automated system is
available
(BRAHMS MR-proADM KRYPTOR; BRAHIV1S GmbH, Hennigsdorf, Germany) (Caruhel
et aL 2009. Clin Bloc/tern 42(7-8):725-8). As these peptides are generated in
a stoichiometric
ratio from the same precursor, their plasma levels are correlated to a certain
extent.
The generation of anti-ADM antibodies is known in the art (e.g. W02013072512
and
W02019057992 incorporated herein by reference).
Another embodiment of the invention relates to an anti-ADM antibody or an anti-
ADM
antibody fragment for use in treatment of shock in a subject that either runs
into shock or that
has developed shock, wherein said antibody or fragment or scaffold binds to
the N-terminal
part (aa 1-21) of mature ADM: YRQSMNNFQGLRSFGCRFGTC (SEQ ID No. 37).
In another preferred embodiment said anti-ADM antibody or an anti-ADM antibody
fragment
recognizes and binds to the N-terminal end (aal) of ADM. N-terminal end means
that the
amino acid 1, that is "Y" of SEQ ID No. 34 and 35 is mandatory for antibody
binding. Said
antibody or fragment or non-Ig scaffold would neither bind N-terminal extended
nor N-
terminally modified ADM nor N-terminally degraded ADM.
Another embodiment of the invention relates to an anti-ADM antibody or an anti-
ADM
antibody fragment for use in treatment of shock in a subject that either runs
into shock or that
has developed shock, wherein said antibody or fragment is a human monoclonal
antibody or
fragment that binds to the N-terminal region (aa 1-21) of mature ADM (SEQ ID
No. 37) or an
antibody fragment thereof wherein the heavy chain comprises the sequences:
CA 03148275 2022-2-16

WO 2021/038078 16
PCT/EP2020/074134
GYTFSRYVV (CDR1: SEQ ID NO: 38), ILPGSGST (CDR2: SEQ ID NO: 39),
TEGYEYDGFDY (CDR3: SEQ ID NO: 40) and
wherein the light chain comprises the sequences:
QSIVYSNGNTY (CDR1: SEQ ID NO: 41), RVS (CDR2), FQGSHIPYT (CDR3: SEQ ID
NO: 42).
Another embodiment of the invention relates to an anti-ADM antibody or an anti-
ADM
antibody fragment for use in treatment of shock in a subject that either runs
into shock or that
has developed shock, wherein said antibody is a monoclonal antibody comprising
the
io following sequence as a heavy chain:
QVQLVQSGAEVKICPGS SVKVSCKASGYTFSRYWIEWVRQAPGQGLEWIGEILPGSG
STNYNQKFQGRVTITADTSTSTAYMELSSLRSEDTAVYYCTEGYEYDGFDYWGQGT
TVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVICDYFPEPVTVSWNSGALTSGVHT
FPAVLQSSGLYSLS S VVTVP S S SL GT QTYICNVNHICP SNTKVDICICVEPICSCDKTHTCP
IS PCPAPELLGGPSVFLFPPICPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEV
HNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGICEYKCKVSNKALPAPIEKTISKAK
GQPREPQVYTLPP SRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL
D SDGSFFLYSKLTVDKSRWQQGNVF SC S VIVIHEALHNHYTQK SL SLSPGK (SEQ ID
NO: 43)
20 and werein said monoclonal antibody is comprising the following sequence
as light chain:
DVVLTQSPLSLPVTLGQPASISCRSSOSIVYSNGNTYLEWYLQRPGQSPRLLIYRVSNR
FSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFQGSHIPYTYGGGTKLEIICRTVAA
PSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWICVDNALQSGNSQESVTEQDSK
DSTYSLSSTLTLSKADYEICHKVYACEVTHQGLSSPVTKSFNRGEC (SEQ ID No. 44).
Endothelial dysfunction can result from and/or contribute to several disease
processes, as
occurs in shock, especially septic shock. From preclinical experiments in
models of sepsis/
septic shock it is known that administration of the anti-ADM antibody induces
an increase of
the plasma bio-ADM concentration and that this coincides with an increased
survival rate
(Struck et al. 2011 Intensive Care Med Exp 1(1):22). The mechanism underlying
this effect
has been described in Geven et al. 2018 (Geven et al. 2018. SHOCK 50 (2): 132-
140).
Briefly, the antibody, when administered i.v., due to its size cannot cross
the endothelial
barrier into the interstitium, but remains in the blood circulation. In
contrast, ADM, as a small
CA 03148275 2022-2-16

WO 2021/038078 17
PCT/EP2020/074134
peptide, can freely diffuse across the endothelial barrier. Thus, the
antibody, when
administered in a vast molar excess over the endogenous ADM, binds virtually
all ADM in
the plasma and, as a simple consequence of reaching binding equilibrium, leads
to a
translocation of ADM from the interstitium to the blood circulation.
Interstitially located
ADM can bind to vascular smooth muscle cells and induces relaxation resulting
in
vasodilation. This is reduced by administration of the antibody. On the other
hand, ADM in
plasma binds to endothelial cells and thereby stabilizes or even restores
vascular integrity.
Thus, this function is strengthened, when plasma ADM levels increase as a
consequence of
administration of the antibody, which is a non-neutralizing antibody. Finally,
binding of the
io antibody to ADM reduces the proteolytic decay of ADM Taken
together, the anti-ADM
antibody Adrecizumab may restore endothelial function in shock e.g. in septic
shock.
If said level of Pro-Adrenomedullin or fragments thereof is above a certain
threshold level the
anti-ADM antibody or anti-ADM antibody fragment binding to ADM is administered
as
therapy.
This means in a specific embodiment subject matter of the present invention
said anti-ADM
antibody or anti-ADM antibody fragment binding to ADM is for therapy, wherein
a sample of
bodily fluid taken from said patients exhibits an elevated level of proADM
and/or fragments
thereof having at least 5 amino acids above a certain threshold. Thus, the
diagnostic method
using said proADM and/or fragments serves as companion diagnostic method.
In a specific embodiment of the invention the threshold is within a threshold
range for plasma
ADM-NH2 that is between 50 and 250 pg/ml, preferably between 55 and 200 pg/ml,
even
more preferred between 60 and 150 pg/ml, even more preferred beween 65 and 100
pg/ml,
most preferred a threshold of 70 pg/ml is applied.
In a specific embodiment of the invention the threshold is within a threshold
range for plasma
MR-proADM that is between 0.5 and 3 nmol/L, preferably between 0.6 and 2
mnol/L, even
more preferred between 0,7 and 1 ng/mL, most preferred a threshold of 0.8
nmol/L is applied.
In a specific embodiment of the invention the threshold is within a threshold
range for plasma
CT-proADM that is between 85 and 500 pmol/L, preferably between 90 and 350
pmol/L,
CA 03148275 2022-2-16

WO 2021/038078 18
PCT/EP2020/074134
even more preferred between 95 and 250 pmol/L, even more preferred between 100
and 200
pmol/L, most preferred a threshold of 150 pmoUL is applied.
The ADM-NH2 levels of the present invention or proADM levels or fragments
thereof,
respectively, have been determined with the described ADM-NH2 assay, as
outlined in the
examples (or proADM or fragments thereof assays, respectively) (Weber et al.
2017. JALM
2(2):1-4). The DPP3 levels of the present invention have been determined with
the described
DPP3-assays as outlined in the examples. The mentioned threshold values above
might be
different in other assays, if these have been calibrated differently from the
assay systems used
io in the present invention. Therefore, the mentioned cut-off values above
shall apply for such
differently calibrated assays accordingly, taking into account the differences
in calibration
One possibility of quantifying the difference in calibration is a method
comparison analysis
(correlation) of the assay in question with the respective biomarker assay
used in the present
invention by measuring the respective biomarker (e.g. bio-ADM, DPP3) in
samples using
is both methods. Another possibility is to determine with the assay in
question, given this test
has sufficient analytical sensitivity, the median biomarker level of a
representative normal
population, compare results with the median biomarker levels as described in
the literature
and recalculate the calibration based on the difference obtained by this
comparison. With the
calibration used in the present invention, samples from normal (healthy)
subjects have been
20 measured: median plasma bio-ADM (mature ADM-NH2) was 24.7 pg/ml, the
lowest value 11
pg/ml and the 996 percentile 43 pg/ml Marino et at 2014. Critical Care
18:R34). With the
calibration used in the present invention, samples from 5,400 normal (healthy)
subjects
(swedish single-center prospective population-based Study (IvIPP-RES)) have
been measured:
median (interquartile range) plasma DPP3 was 14.5 ng/m1 (11.3 ng/ml ¨ 19
ng/rnl).
The plasma median MR-proADM concentration in normal (healthy) subjects was
0.41
(interquartile range 0.23 - 0.64) nmoUL (Smith et at 2009. Clin Chem 55:1593-
1595) using
the automated sandwich fluorescence assay for the detection of MR-proADM as
described in
Caruhel et al. (Caruhel et at 2009. Clin Biochem 42:725-8).
The plasma median concentration of CT-proADM in normal healthy subjects
(n=200) was
77.6 pmol/L (min 46.6 pmol/L, max 136.2 pmol/L) and the 95% percentile was
113.8 pmol/L
(EP 2 111 552B1).
CA 03148275 2022-2-16

WO 2021/038078 19
PCT/EP2020/074134
In a specific embodiment of the invention a threshold for plasma ADM-NH2 is
the 5fold
median concentration, preferably the 4fo1d median concentration, more
preferred the 3fold
median concentration, most preferred the 2fo1d median concentration of a
normal healthy
population.
In a specific embodiment of the invention a threshold for plasma MR-proADM is
the 5fold
median concentration, preferably the 4fo1d median concentration, more
preferred the 3fo1d
median concentration, most preferred the 2fo1d median concentration of a
normal healthy
population.
Io
In a specific embodiment of the invention a threshold for plasma CT-proADM is
the 5fold
median concentration, preferably the 4fold median concentration, more
preferred the 3fold
median concentration, most preferred the 2fo1d median concentration of a
normal healthy
population.
In embodiments of the present invention, the anti-ADM antibody or an anti-ADM
antibody
fragment for use in a treatment of a patient in need thereof, may be
administered in a dose of
at least 0.5 mg / Kg body weight, particularly at least 1.0 mg/kg body weight,
more
particularly, from 1.0 to 20.0 mg/kg body weight, e.g., from 2.0 to 10 mg/kg
body weight,
from 2.0 to 8.0 mg/kg body weight, or from 2.0 to 5.0 mg/kg body weight.
One embodiment of the invention relates to a vasopressor for use in therapy of
shock in a
subject that either runs into shock or that has developed shock, wherein said
subject has a
level of DPP3 in a sample of bodily fluid of said subject that is below a
predetermined
threshold, when determined by any of the methods of predicting or diagnosing a
refractory
shock according to the present invention.
Another embodiment of the invention relates to a vasopressor for use in
therapy of shock in a
subject that either runs into shock or that has developed shock, wherein said
vasopressor is
selected from the group comprising dopamine, norepinephrine, a norepinephrine
equivalent,
epinephrine, phenylephrine and vasopressin.
CA 03148275 2022-2-16

WO 2021/038078 20
PCT/EP2020/074134
Another specific embodiment of the invention relates to a vasopressor for use
in therapy of
shock in a subject that either runs into shock or that has developed shock,
wherein said
vasopressor is administered to said subject in a pharmaceutical formulation.
Another embodiment of the invention relates to a vasopressor for use in
therapy of shock in a
subject that either runs into shock or that has developed shock, wherein said
subject has a
blood pressure of equal or less than 65 mm Hg.
Another embodiment of the invention relates to an inhibitor of the activity of
DPP3 for use in
io therapy of shock in a subject that either runs into shock or that has
developed shock, wherein
said subject has a level of DPP3 in a sample of bodily fluid of said subject
that is above a
predetermined threshold when determined by a method described in the
aforementioned
embodiments.
IS Another specific embodiment of the invention relates to an inhibitor of
the activity of DPP3
for use in therapy of shock in a subject That either runs into shock or that
has developed shock,
wherein the inhibitor of the activity of DPP3 is selected from the group
comprising anti-DPP3
antibody or anti-DPP3 antibody fragment or anti-DPP3 non-Ig scaffold.
20 One embodiment of the invention relates to an inhibitor of the activity
of DPP3 for use in
therapy of shock in a subject that either runs into shock or that has
developed shock, wherein
said inhibitor has a minimum binding affinity to DPP3 of equal or less than
104 M.
Another embodiment of the invention relates to an inhibitor of the activity of
DPP3 for use in
25 therapy of shock in a subject that either runs into shock or that has
developed shock, wherein
the inhibitor of the activity of DPP3 is an antibody.
Another specific embodiment of the invention relates to an inhibitor of the
activity of DPP3
for use in therapy of shock in a subject that either runs into shock or that
has developed shock,
30 wherein said inhibitor is an antibody that is monoclonal_
Another embodiment of the invention relates to an inhibitor of the activity of
DPP3 for use in
therapy of shock in a subject that either runs into shock or that has
developed shock, wherein
said inhibitor is a monoclonal antibody, wherein the complementaiity
determining regions
35 (CDR's) in the heavy chain comprises the sequences: SEQ NO.: 7, SEQ ID
NO.: 8 and/ or
CA 03148275 2022-2-16

WO 2021/038078 21
PCT/EP2020/074134
SEQ ID NO.: 9 and the complementarily determining regions (CDR's) in the light
chain
comprises the sequences: SEQ ID NO: 10, KVS and/or SEQ ID NO.: 11.
Another embodiment of the invention relates to an inhibitor of the activity of
DPP3 for use in
therapy of shock in a subject that either runs into shock or that has
developed shock, wherein
said inhibitor is a humanized monoclonal antibody or humanized monoclonal
antibody
fragment, wherein the heavy chain comprises the sequence: SEQ ID NO.: 12 and
wherein the
light chain comprises the sequence. SEQ ID NO: 13.
io Said inhibitor of the activity of DPP3 maybe administered by inhalation.
One embodiment of the invention relates to an inhibitor of the activity of
DPP3 for use in
therapy of shock in a subject that either runs into shock or that has
developed shock, wherein
said inhibitor is administered in combination with an Angiotensin-Receptor-
Agonist and/or
precursors thereof
Another embodiment of the invention relates to an inhibitor of the activity of
DPP3 for use in
therapy of shock in a subject that either runs into shock or that has
developed shock, wherein
said Angiotensin-Receptor-Agonist and/or precursors thereof is selected from
the group
comprising angiotensin I, angiotensin II, angiotensin III, angiotensin IV, in
particular
angiotensin
Another embodiment of the invention relates to an Angiotensin-Receptor-
Agonists and/ or
precursors thereof and/or inhibitors of the DPP3 activity for use in the
treatment of shock in a
subject that either runs into shock or that has developed shock, wherein the
treatment with
said Angiotensin-Receptor-Agonists and/ or precursors thereof and/or
inhibitors of the DPP3
activity is initiated and/or continued when the level of DPP3 in a sample of
said subject is
above a certain threshold and/or wherein a treatment with vasopressors is
withheld and/ or
terminated if said determined level of DPP3 is above said predetermined
threshold.
Another specific embodiment of the invention relates to a vasopressor for use
in the treatment
of shock in a subject that either runs into shock or that has developed shock,
wherein the
treatment with said vasopressor is initiated and/or continued when the level
of DPP3 in a
sample of bodily fluid of said subject is below a certain threshold and/or
wherein a treatment
CA 03148275 2022-2-16

WO 2021/038078 22
PCT/EP2020/074134
with Angiotensin-Receptor-Agonists and/ or precursors thereof and/or
inhibitors of the DPP3
activity is withheld and/ or terminated if the said determined level of DPP3
is below said
predetermined threshold.
Another specific embodiment of the invention relates to a vasopressor for use
in the treatment
of shock in a subject that either runs into shock or that has developed shock,
wherein the
treatment with said vasopressor is initiated and/or continued when the level
of DPP3 in a
sample of bodily fluid of said subject is below a certain threshold and/or
wherein a treatment
with Angiotensin-Receptor-Agonists and/ or precursors thereof and/or
inhibitors of the DPP3
m activity is withheld and/ or terminated if the said determined level of
DPP3 is below said
predetermined threshold and wherein the level of Pro-adrenomedullin or
fragments thereof is
determined and wherein treatment with said anti-ADM antibody or anti-ADM
antibody
fragment is initiated and/or continued when the level of Pro-adrenomedullin or
fragments
thereof in said sample is above a certain threshold and/or wherein a treatment
with an anti-
ADM antibody or anti-ADM antibody fragment is withheld and/ or terminated if
the said
determined level of Pro-adrenomedullin or fragments thereof is below said
predetermined
threshold.
Another embodiment of the invention relates to Angiotensin-Receptor-Agonists
and/ or
precursors thereof and/or inhibitors of the DPP3 activity for use in the
treatment of shock in a
subject that either runs into shock or that has developed shock, wherein the
treatment with
said vasopressor is initiated and/or continued when the level of DPP3 in a
sample of bodily
fluid of said subject is below a certain threshold and/or wherein a treatment
with
Angiotensin-Receptor-Agonists and/ or precursors thereof and/or inhibitors of
the DPP3
activity is withheld and/ or terminated if the said determined level of DPP3
is below said
predetermined threshold, and wherein the level of Pro-adrenomedullin or
fragments thereof is
determined and wherein treatment with said anti-ADM antibody or anti-ADM
antibody
fragment is initiated and/or continued when the level of Pro-adrenomedullin or
fragments
thereof in said sample is above a certain threshold and/or wherein a treatment
with an anti-
ADM antibody or anti-ADM antibody fragment is withheld and/ or terminated if
the said
determined level of Pro-adrenomedullin or fragments thereof is below said
predetermined
threshold.
CA 03148275 2022-2-16

WO 2021/038078 23
PCT/EP2020/074134
Another specific embodiment of the invention relates to anti-ADM antibody or
anti-ADM
antibody fragment for use in the treatment of shock in a subject that either
runs into shock or
that has developed shock, wherein treatment with said anti-ADM antibody or
anti-ADM
antibody and/or Angiotensin-Receptor-Agonists and/ or precursors thereof
and/or inhibitors
of the DPP3 activity is initiated and/or continued if the level of Pro-
adrenomedullin or
fragments thereof in said sample is above a certain threshold and said
determined level of
DPP3 is above said predetermined threshold of DPP3.
In case the level of of Pro-adrenomedullin or fragments thereof in said sample
is determined
io and the level of DPP3 in said sample is determined said determination
maybe conducted by a
point-of-care device that provides both: a test for determining level of Pro-
adrenomedullin or
fragments thereof and a test for determining the level of DPP3 in said sample.
Beginning of treatment with an angiotensin-receptor-agonist and/ or a
precursor thereof is
indicated when a patient has an amount of DPP3 protein and/or DPP3 activity in
a sample of
bodily fluid That is above a predetermined threshold level.
One embodiment of the invention relates to a method of treatment of shock in a
subject that
either runs into shock or that has developed shock, the method comprising
administering
vasopressor to said subject, wherein said subject has a level of DPP3 in a
sample of bodily
fluid of said subject that is below a predetermined threshold, when determined
by a method
described in previous embodiments.
Another embodiment of the invention relates to a method of treatment of shock
in a subject
that either runs into shock or that has developed shock, the method comprising
administering
vasopressor to said subject, wherein said vasopressor is selected from the
group comprising
dopamine, norepinephrine, a norepinephrine equivalent, epinephrine,
phenylephrine and
vasopressin.
A specific embodiment of the invention relates to a method of treatment of
shock in a subject
that either runs into shock or that has developed shock, the method comprising
administering
vasopressor to said subject, wherein said vasopressor is administered to said
subject in a
pharmaceutical formulation.
CA 03148275 2022-2-16

WO 2021/038078 24
PCT/EP2020/074134
Another embodiment of the invention relates to a method of treatment of shock
in a subject
that either runs into shock or that has developed shock, the method comprising
administering
vasopressor to said subject, wherein said subject has a blood pressure of
equal or less than 65
min Hg.
One embodiment of the invention relates to a method of treatment of shock in a
subject that
either runs into shock or that has developed shock, the method comprising
administering
inhibitor of DPP3 activity to said subject, wherein said subject has a level
of DPP3 in a
sample of bodily fluid of said subject that is above a predetermined threshold
when
io determined by a method described in previous embodiments.
Another embodiment of the invention relates to a method of treatment of shock
in a subject
that either runs into shock or that has developed shock, the method comprising
administering
inhibitor of DPP3 activity to said subject, wherein the inhibitor of the
activity of DPP3 is
selected from the group comprising anti-DPP3 antibody or anti-DPP3 antibody
fragment or
anti-DPP3 non-Ig scaffold.
Another specific embodiment of the invention relates to a method of treatment
of shock in a
subject that either runs into shock or that has developed shock, the method
comprising
administering inhibitor of DPP3 activity to said subject, wherein said
inhibitor has a minimum
binding affinity to DPP3 of equal or less than le M.
Another embodiment of the invention relates to a method of treatment of shock
in a subject
that either runs into shock or that has developed shock, the method comprising
administering
inhibitor of DPP3 activity to said subject, wherein the inhibitor of the
activity of DPP3 is an
antibody.
One embodiment of the invention relates to a method of treatment of shock in a
subject that
either runs into shock or that has developed shock, the method comprising
administering
inhibitor of DPP3 activity to said subject, wherein said inhibitor is an
antibody that is
monoclonal.
One specific embodiment of the invention relates to a method of treatment of
shock in a
subject that either tuns into shock or that has developed shock, the method
comprising
CA 03148275 2022-2-16

WO 2021/038078 25
PCT/EP2020/074134
administering inhibitor of DPP3 activity to said subject, wherein said
inhibitor is a
monoclonal antibody, wherein the complementarity determining regions (CDR's)
in the
heavy chain comprises the sequences: SEQ ID NO.: 7, SEQ ID NO.: 8 and/ or SEQ
ID NO.: 9
and the complementarity determining regions (CDR's) in the light chain
comprises the
sequences: SEQ lID NO.: 10, KVS and/or SEQ ID NO.: 11.
Another embodiment of the invention relates to a method of treatment of shock
in a subject
that either runs into shock or that has developed shock, the method comprising
administering
inhibitor of DPP3 activity to said subject, wherein said inhibitor is a
humanized monoclonal
io antibody or humanized monoclonal antibody fragment, wherein the heavy chain
comprises
the sequence: SEQ ID NO.: 12 and wherein the light chain comprises the
sequence: SEQ ID
NO.: 13.
Another specific embodiment of the invention relates to a method of treatment
of shock in a
subject that either runs into shock or that has developed shock, the method
comprising
administering inhibitor of DPP3 activity to said subject, wherein said
inhibitor is administered
in combination with an Angiotensin-Receptor-Agonist and/or precursors thereof
Another embodiment of the invention relates to a method of treatment of shock
in a subject
that either runs into shock or that has developed shock, the method comprising
administering
inhibitor of DPP3 activity to said subject, wherein said Angiotensin-Receptor-
Agonist and/or
precursors thereof is selected from the group comprising angiotensin I,
angiotensin II,
angiotensin Ill, angiotensin IV, in particular angiotensin II.
Another embodiment of the invention relates to a method of treatment of shock
in a subject
that either runs into shock or that has developed shock, the method comprising
administering
Angiotensin-Receptor-Agonists and/or precursors thereof and/or inhibitor of
DPP3 activity to
said subject, wherein the treatment with said Angiotensin-Receptor-Agonists
and/or
precursors thereof and/or inhibitors of the DPP3 activity is initiated and/or
continued when
the level of DPP3 in a sample of said subject is above a certain threshold
and/or wherein a
treatment with vasopressors is withheld and/ or terminated if said determined
level of DPP3 is
above said predetermined threshold.
CA 03148275 2022-2-16

WO 2021/038078 26
PCT/EP2020/074134
One embodiment of the invention relates to a method of treatment of shock in a
subject that
either runs into shock or that has developed shock, the method comprising
administering
vasopressor to said subject, wherein the treatment with said vasopressors is
initiated and/or
continued when the level of DPP3 in a sample of bodily fluid of said subject
is below a
certain threshold and/or wherein a treatment with Angiotensin-Receptor-
Agonists and/or
precursors thereof and/or inhibitors of the DPP3 activity is withheld and/ or
terminated if the
said determined level of DPP3 is below said predetermined threshold.
One embodiment of the invention relates to a method of treatment of shock in a
subject that
io either runs into shock or that has developed shock, the method comprising
administering
Angiotensin-Receptor-Agonists and/ or precursors thereof and/or inhibitor of
DPP3 activity
to said subject, wherein the treatment with said Angiotensin-Receptor-Agonists
and/or
precursors thereof and/or inhibitors of the DPP3 activity is initiated and/or
continued when
the level of DPP3 in a sample of said subject is above a certain threshold
and/or wherein a
treatment with vasopressors is withheld and/ or terminated if said determined
level of DPP3 is
above said predetermined threshold and wherein the level of Pro-adrenomedullin
or fragments
thereof is determined and wherein treatment with said anti-ADM antibody or
anti-ADM
antibody fragment is initiated and/or continued when the level of Pro-
adrenomedullin or
fragments thereof in said sample is above a certain threshold and/or wherein a
treatment with
an anti-ADM antibody or anti-ADM antibody fragment is withheld and/ or
terminated if the
said determined level of Pro-adrenomedullin or fragments thereof is below said

predetermined threshold.
One specific embodiment of the invention relates to a method of treatment of
shock in a
subject that either runs into shock or that has developed shock., the method
comprising
administering vasopressor to said subject, wherein the treatment with said
vasopressors is
initiated and/or continued when the level of DPP3 in a sample of bodily fluid
of said subject
is below a certain threshold and/or wherein a treatment with Angiotensin-
Receptor-Agonists
and/or precursors thereof and/or inhibitors of the DPP3 activity is withheld
and/ or terminated
if the said determined level of DPP3 is below said predetermined threshold and
wherein the
level of Pro-adrenomedullin or fragments thereof is determined and wherein
treatment with
said anti-ADM antibody or anti-ADM antibody fragment is initiated and/or
continued when
the level of Pro-adrenomedullin or fragments thereof in said sample is above a
certain
threshold and/or wherein a treatment with an anti-ADM antibody or anti-ADM
antibody
CA 03148275 2022-2-16

WO 2021/038078 27
PCT/EP2020/074134
fragment is withheld and/ or terminated if the said determined level of Pro-
adrenomedullin or
fragments thereof is below said predetermined threshold.
Another embodiment of the invention relates to a method of treatment of shock
in a subject
that either runs into shock or that has developed shock, the method comprising
administering
anti-ADM antibody or anti-ADM antibody fragment to said subject, wherein the
level of Pro-
adrenomedullin or fragments thereof is determined and wherein treatment with
said anti-
ADM antibody or anti-ADM antibody fragment is initiated and/or continued when
the level
of Pro-adrenomedullin or fragments thereof in said sample is above a certain
threshold and/or
io wherein a treatment with an anti-ADM antibody or anti-ADM antibody
fragment is withheld
and/ or terminated if the said determined level of Pro-adrenomedullin or
fragments thereof is
below said predetermined threshold and wherein the treatment with Angiotensin-
Receptor-
Agonists and/or precursors thereof and/or inhibitors of the DPP3 activity is
initiated and/or
continued when the level of DPP3 in a sample of said subject is above a
certain threshold
and/or wherein a treatment with vasopressors is withheld and/ or terminated if
said
determined level of DPP3 is above said predetermined threshold
Another specific embodiment of the invention relates to a method of treatment
of shock in a
subject that either runs into shock or that has developed shock, the method
comprising
administering anti-ADM antibody or anti-ADM antibody fragment to said subject,
wherein
treatment with said anti-ADM antibody or anti-ADM antibody and/or Angiotensin-
Receptor-
Agonists and/ or precursors thereof and/or inhibitors of the DPP3 activity is
initiated and/or
continued if the level of Pro-adrenomedullin or fragments thereof in said
sample is above a
certain threshold and said determined level of DPP3 is above said
predetermined threshold of
DPP3.
An agonist means a chemical that binds to a receptor and activates the
receptor to produce a
biological response. Receptors can be activated by either endogenous agonists
(such as
hormones and neurotransmitters) or exogenous agonists (such as drugs),
resulting in a
biological response. Full agonists bind to and activate a receptor with the
maximum response
that an agonist can elicit at the receptor. Partial agonists are drugs that
bind to and activate a
given receptor, but have only partial efficacy at the receptor relative to a
full agonist. Potency
is the amount of agonist needed to elicit a desired response. The potency of
an agonist is
inversely related to its EC50 value The EC50 can be measured for a given
agonist by
CA 03148275 2022-2-16

WO 2021/038078 28
PCT/EP2020/074134
determining the concentration of agonist needed to elicit half of the maximum
biological
response of the agonist. The EC50 value is useful for comparing the potency of
drugs with
similar efficacies producing physiologically similar effects. The smaller the
EC50 value, the
greater the potency of the agonist, the lower the concentration of drug that
is required to elicit
the maximum biological response.
The treatment with an angiotensin-receptor-agonist and/or a precursor thereof
may be
continued as long as the patient has an amount of DPP3 protein and/or DPP3
activity in a
sample of bodily fluid that is above a predetermined threshold level.
io
The amount of DPP3 protein and/ or the DPP3 activity is measured at least
every 24 hours,
preferred every 12 hours, more preferred every 8 hours, even more preferred
every 6 hours,
even more preferred every 4 hours, even more preferred every 2 hours, even
more preferred
every hour, most preferred every 30 minutes.
The treatment with an angiotensin-receptor-agonist and/ or a precursor thereof
may be
discontinued when the patient has an amount of DPP3 protein and/or DPP3
activity in a
sample of bodily fluid that is below a predetermined threshold level.
Angiotensin Therapeutics:
Angiotensin I, also called proangiotensin, is formed by the action of renin on

angiotensinogen. Renin cleaves the peptide bond between the leucine (Leu) and
valine (Val)
residues on angiotensinogen, creating the decapeptide (ten amino acid) (des-
Asp) angiotensin
I Angiotensin I is converted to angiotensin II through removal of two C-
terminal residues by
the enzyme angiotensin-converting enzyme (ACE), primarily through ACE within
the lung
(but also present in endothelial cells, kidney epithelial cells, and the
brain).
Angiotensin II, angiotensin III, and angiotensin IV are peptide hormones
naturally produced
by the body that regulates blood pressure via vasoconstriction and sodium
reabsorption
Hemodynamic effects of angiotensin II administration have been the subject of
numerous
clinical studies, demonstrating significant effects on systemic and renal
blood flow (Harrison-
Bernard, L.M. The renal renin-angiotensin system. Adv Physiol Ethic, 33(4):
270 (2009)).
CA 03148275 2022-2-16

WO 2021/038078 29
PCT/EP2020/074134
Angiotensin II is a hormone produced by the renin angiotensin aldosterone
system (RAAS)
that modulates blood pressure via regulation of vascular smooth muscle tone
and extracellular
fluid homeostasis. Angiotensin II mediates its effects on the vasculature by
inducing
vasoconstriction and sodium retention, and so is the target of many therapies
for hypertension.
In addition to its systemic effects, angiotensin II has a pronounced effect on
the efferent
arterioles of the kidney, maintaining glomerular filtration when blood flow is
decreased
Angiotensin II also regulates sodium reabsorption in the kidney by stimulating
Na-E/H+
exchangers in the proximal tubule and inducing the release of aldosterone and
vasopressin
(Harrison-Bernard 2009. The renal renin-angiotensin system. Ads, Physiol Edw.
33(4):270).
The angiotensin II therapeutic that may be used in the compositions and
methods of this
disclosure may be Asp-Arg-Val-Tyr-Ile-His-Pro-Phe (SEQ ID NO: 13), also called

5-isoleucine angiotensin II. SEQ ID NO: 13 is an octa-peptide naturally
present in humans
and other species, such as equines, hogs, etc. Isoleucine may be substituted
by valine to result
in 5-vaiine angiotensin IL, Asp-Arg-Val-Tyr-Val-His-Pro-Phe (SEQ ID NO: 14).
Other
angiotensin II analogues such as [Asni-Phel-angiotensin II (SEQ ID NO: 15),
hexapeptide
Val-Tyr-Ile-His-Pro-Phe (SEQ ID NO: 16), nonapeptide Asn-Arg-Val-Tyr-Tyr-Val-
His-Pro-
Phe (SEQ ID NO: 17), [As& Ile5 Ilel-angiotensin II (SEQ ID NO: 18), [Asnl -
1Ie5- Ala8]-
angiotensin II (SEQ ID NO: 19), and [Asni-diiodoTyr4-Ile5-angiotensin II (SEQ
ID NO: 20)
may also be used. Angiotensin II may be synthesized, for example, by solid
phase peptide
synthesis to incorporate modifications, such as C -terminal amidation_ The
term "angiotensin
II", without further specificity, is intended to refer to any of these various
forms, as well as
combinations thereof.
In some aspects, a composition comprising angiotensin II may be selected from
5-valine
angiotensin II, 5-valine angiotensin II amide, 5-L-isoleucine angiotensin II,
and
5-L-isoleucine angiotensin II amide, or a pharmaceutically acceptable salt
thereof, preferably
manufactured under current good manufacturing conditions (cGMP). In some
aspects, the
composition may include different forms of angiotensin II in different
percentages, e.g., a
mixture of hexapeptide and nonapeptide angiotensin II. The composition
comprising
angiotensin II may be suitable for parenteral administration, e.g., for
injection or intravenous
infusion.
CA 03148275 2022-2-16

WO 2021/038078 30
PCT/EP2020/074134
The sequence of angiotensin II used in the compositions and methods disclosed
herein may be
homologous to the sequences of angiotensin II described above. In certain
aspects, the
invention includes isolated, synthetic, or recombinant amino acid sequences
that are at least
80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 13, 14, 15,
16, 17,
18, 19 and/or 20. Any such variant sequences may be used in place of an
angiotensin II as
described in the preceding paragraph.
Sequence identity for all sequences is determined according to the following
method: Percent
identity is calculated by multiplying the number of matches in the pair by 100
and dividing by
the length of the aligned region, including gaps (Percent Identity = [Matches
x 1001/Length of
to aligned region [with gaps").
Angiotensin III is a metabolite of angiotensin II with approximately 40% of
the activity of
angiotensin II. An angiotensin III therapeutic that may be used for in the
compositions and
methods of this disclosure may be Arg-Val-Tyr-Ile-His-Pro-Phe (SEQ ID NO: 21).
SEQ ID
NO: 21 is an hepta-peptide naturally present in humans and other species, such
as equines,
hogs, etc. Isoleucine may be substituted by valine to result in Arg-Val-Tyr-
Val-His-Pro-Phe
(SEQ ID NO: 22). Other angiotensin III analogues such as [Phei-angiotensin III
(SEQ ID
NO: 23), [Ile4-Ala7]-angiotensin III (SEQ ID NO: 24), and [diiodoTyr34161-
angiotensin III
(SEQ ID NO: 25) may also be used. Angiotensin III may be synthesized, for
example, by
solid phase peptide synthesis to incorporate modifications, such as C-terminal
amidation. The
term "angiotensin III", without further specificity, is intended to refer to
any of these various
forms, as well as combinations thereof.
In some aspects, a composition comprising angiotensin III may be selected from
4-valine
angiotensin III, 4-valine angiotensin III amide, 4-L-isoleucine angiotensin
III, and
4-L-isoleueine angiotensin III amide, or a pharmaceutically acceptable salt
thereof, preferably
manufactured under current good manufacturing conditions (cGMP). A composition

comprising angiotensin III may be suitable for parenteral administration,
e.g., for injection or
intravenous infusion.
The sequence of angiotensin III used in the compositions and methods disclosed
herein may
be homologous to the sequences of angiotensin III described above. In certain
aspects, the
invention includes isolated, synthetic, or recombinant amino acid sequences
that are at least
80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 21, 22, 23,
24
CA 03148275 2022-2-16

WO 2021/038078 31
PCT/EP2020/074134
and/or 25. Any such variant sequences may be used in place of an angiotensin
II as described
in the preceding paragraph.
Angiotensin IV is a metabolite of angiotensin III with less activity than
angiotensin IL An
angiotensin IV therapeutic that may be used for in the compositions and
methods of this
disclosure may be Val-Tyr-Ile-His-Pro-Phe (SEQ ID NO: 26). SEQ ID NO: 26 is a
hexa-
peptide naturally present in humans and other species. Isoleucine may be
substituted by
valine to result in Val-Tyr-Val-His-Pro-Phe (SEQ ID NO: 27). Other angiotensin
IV
analogues such as Whe2kangiotensin III (SEQ ID NO: 28), [Ile3-A1a6]-
angiotensin IV (SEQ
io ID NO: 29), and [diiodoTyr2-Ile3]-angiotensin IV (SEQ ID NO: 30) may also
be used.
Angiotensin IV may be synthesized, for example, by solid phase peptide
synthesis to
incorporate modifications, such as C-terminal amidation. The term "angiotensin
IV", without
further specificity, is intended to refer to any of these various forms, as
well as combinations
thereof
In some aspects, a composition comprising angiotensin IV may be selected from
3-valine
angiotensin IV, 3-valine angiotensin IV amide, 3-L-isoleucine angiotensin IV,
and
3-L-isoieucine angiotensin IV amide, or a pharmaceutically acceptable salt
thereof, preferably
manufactured under current good manufacturing conditions (cGMP). A composition

comprising angiotensin IV may be suitable for parenteral administration, e.g.,
for injection or
intravenous infusion.
The sequence of angiotensin IV used in the compositions and methods disclosed
herein may
be homologous to the sequences of angiotensin IV described above. In certain
aspects, the
invention includes isolated, synthetic, or recombinant amino acid sequences
that are at least
80%, 85%, 90%, 95%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 25, 26, 27,
28, 29
and/or 30. Any such variant sequences may be used in place of an angiotensin
IV as described
in the preceding paragraph.
An angiotensin II, angiotensin III, or angiotensin IV therapeutic may be used
as any suitable
salt (e.g. acetate), deprotected form, acetylated form, deacetylated form,
and/or prodrug form
of the above-mentioned peptides, including pegylated forms of the peptides or
conjugates as
disclosed in US Patent Publication 2011/0081371 (incorporated by reference).
The term
"prodrug" refers to any precursor compound which is able to generate or to
release the above-
mentioned peptide under physiological conditions. Such prodrugs or precursors
may be larger
CA 03148275 2022-2-16

WO 2021/038078 32
PCT/EP2020/074134
peptides which are selectively cleaved in order to form the peptide of the
invention. For
example, in some aspects, the prodrug or precursor may be angiotensinogen,
angiotensin I, or
its homologues that may result in angiotensin II by the action of certain
endogenous or
exogenous enzymes. Further prodrugs or precursors include peptides with
protected amino
acids, e.g., having protecting groups at one or more carboxylic acid and/or
amino groups.
Suitable protecting groups for amino groups include the benzyloxycarbonyl, t-
butyloxycarbonyl (BOC), fluorenylmethyloxycarbonyl (FMOC), formyl, and acetyl
or acyl
group. Suitable protecting groups for the carboxylic acid group include esters
such as benzyl
esters or t-butyl esters. The present invention also contemplates the use of
angiotensin II,
io angiotensin III, angiotensin IV and/or precursor peptides having amino acid
substitutions,
deletions, additions, the substitutions and additions including the standard D
and L amino
acids and modified amino acids, such as, for example, amidated and acetylated
amino acids,
wherein the therapeutic activity of the base peptide sequence is maintained at
a
pharmacologically useful level.
In a preferred embodiment the angiotensin II is angiotensin II acetate.
Angiotensin II acetate
is L-Aspartyl-L-arginyl-L-valyl-Ltyrosyl-L-i soleucyl-L-
histidyl-L-prolyl-L-phenylaIanine,
acetate salt. The counter ion acetate is present in a non-stoichiometric
ratio. The molecular
formula of angiotensin II acetate is C50H7INI3012 = (C2H402)n; (n=number of
acetate
molecules; theoretical n = 3) with an average molecular weight of 1046.2 (as
free base).
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
thereof for use in the treatment of a disease in a subject, wherein said
subject has an amount
of DPP3 protein and/or DPP3 activity in a sample of bodily fluid that is above
a
predetermined threshold, wherein the determination of the amount of DPP3
protein and/or
DPP3 activity in a sample of bodily fluid is used as therapy guidance and/or
therapy
monitoring for said treatment with Angiotensin-Receptor-Agonist and/ or a
precursor thereof
In one embodiment of the invention the amount of DPP3 protein and/or DPP3
activity in a
sample of bodily fluid is determined before treatment of the subject with an
angiotensin-
receptor-agonist and/ or a precursor thereof In one embodiment of the
invention the amount
of DPP3 protein and/or DPP3 activity in a sample of bodily fluid is determined
during
treatment of the subject with an angiotensin-receptor-agonist and/ or a
precursor thereof at
least once during treatment, preferably at least twice or preferably at least
once daily during
CA 03148275 2022-2-16

WO 2021/038078 33
PCT/EP2020/074134
treatment. In one embodiment of the invention the amount of DPP3 protein
and/or DPP3
activity in a sample of bodily fluid is determined after treatment of the
subject with an
angiotensin-receptor-agonist and/ or a precursor thereof In one embodiment of
the invention
the amount of DPP3 protein and/or DPP3 activity in a sample of bodily fluid is
determined
before and/or during and/or after treatment of the subject with an angiotensin-
receptor-agonist
and/ or a precursor thereof
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
thereof for use in the treatment ofshock in a subject that either runs into
shock or that has
io developed shock , wherein said subject has an amount of DPP3 protein
and/or DPP3 activity
in a sample of bodily fluid that is above a predetermined threshold, wherein
said sample of
bodily fluid of said subject is selected from whole blood, blood plasma and
blood serum.
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
IS thereof for use in the treatment of shock in a subject that either runs
into shock or that has
developed shock, wherein said Angiotensin-Receptor-Agonist and/ or a precursor
thereof is
administered to said subject in a pharmaceutical formulation.
The term "pharmaceutical formulation" means a pharmaceutical (active)
ingredient in
20 combination with at least one pharmaceutically acceptable excipient.
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
thereof for use in the treatment of shock in a subject that either runs into
shock or that has
developed shock, wherein said Angiotensin-Receptor-Agonist and/ or a precursor
thereof is
25 Angiotensin II and said pharmaceutical formulation is a solution,
preferably a ready-to-use
solution. In another embodiment subject of the present invention is further a
pharmaceutical
formulation according to the present invention wherein said pharmaceutical
formulation is in
a dried state to be reconstituted before use.
30 The pharmaceutical formulation as disclosed herein may also contain
diluents, fillers, salts,
buffers, stabilizers, solubilizers, and other materials well known in the art.
The term
"pharmaceutically acceptable carrier" refers to a non-toxic carrier that may
be administered to
a patient, together with a therapeutically effective substance (such as
angiotensin II) of this
invention, and which does not destroy the pharmacological activity of the
therapeutically
CA 03148275 2022-2-16

WO 2021/038078 34
PCT/EP2020/074134
effective substance. The term "pharmaceutically acceptable" means a non-toxic
material that
does not interfere with the effectiveness of the biological activity of the
active ingredient(s).
The characteristics of the carrier will depend on the route of administration.
The term
"excipient" refers to an additive in a formulation or composition that is not
a pharmaceutically
active ingredient.
The term "pharmaceutical (active) ingredient" means a therapeutic composition
which can be
optionally combined with pharmaceutically acceptable excipients to provide a
pharmaceutical
io formulation or dosage form.
One of skill in the art would appreciate that the choice of any one excipient
may influence the
choice of any other excipient. For example, the choice of a particular
excipient may preclude
the use of one or more additional excipients because the combination of
excipients would
produce undesirable effects. Excipients of the invention may include, but are
not limited to,
co-solvents, solubilizing agents, buffers, pH adjusting agents, bulking
agents, surfactants,
encapsulating agents, tonicity-adjusting agents, stabilizing agents,
protectants, and viscosity
modifiers.
In some aspects, it may be beneficial to include a pharmaceutically acceptable
carrier in the
compositions as dislosed herein.
In some aspects, it may be beneficial to include a solubilizing agent in the
compositions of the
invention. Solubilizing agents may be useful for increasing the solubility of
any of the
components of the formulation or composition, including a therapeutically
effective substance
(e.g., angiotensin II, angiotensin III, or angiotensin IV) or an excipient.
The solubilizing
agents described herein are not intended to constitute an exhaustive list, but
are provided
merely as exemplary solubilizing agents that may be used in the compositions
of the
invention. In certain aspects, solubilizing agents include, but are not
limited to, ethyl alcohol,
tert-butyl alcohol, polyethylene glycol, glycerol, methylparaben,
propylparaben, polyethylene
glycol, polyvinyl pyrrolidone, and any pharmaceutically acceptable salts
and/or combinations
thereof
CA 03148275 2022-2-16

WO 2021/038078 35
PCT/EP2020/074134
In some aspects, it may be beneficial to adjust the pH of the compositions by
including a
pH-adjusting agent in the compositions of the invention. Modifying the pH of a
formulation
or composition may have beneficial effects on, for example, the stability or
solubility of a
therapeutically effective substance, or may be useful in making a formulation
or composition
suitable for parenteral administration, pH-adjusting agents are well known in
the art.
Accordingly, the pH-adjusting agents described herein are not intended to
constitute an
exhaustive list, but are provided merely as exemplary pH-adjusting agents that
may be used in
the compositions of the invention, pH-adjusting agents may include, for
example, acids and
bases. In some aspects, a pH-adjusting agent includes, but is not limited to,
acetic acid,
io hydrochloric acid, phosphoric acid, sodium hydroxide, sodium carbonate, and
combinations
thereof
The pH of the compositions as disclosed herein may be any pH that provides
desirable
properties for the formulation or composition. Desirable properties may
include, for example,
therapeutically effective substance (e.g., angiotensin II, angiotensin III, or
angiotensin IV)
stability, increased therapeutically effective substance retention as compared
to compositions
at other pHs, and improved filtration efficiency. In some aspects, the pH of
the compositions
of the invention may be from about 3.0 to about 9.0, e.g., from about 5.0 to
about 7Ø In
particular aspects, the pH of the compositions of the invention may be
5.5+0.1, 5.6+0.1,
5.7+0.1, 5.8+0.1, 5.9+0.1, 6.0+0.1, 6.1+0.1, 6.2+0.1, 6.3+0.1, 6.4 0.I, or 6.5
-0.1.
In some aspects, it may be beneficial to buffer the pH by including one or
more buffers in the
compositions. In certain aspects, a buffer may have a pKa of, for example,
about 5.5, about
6.0, or about 6.5. One of skill in the art would appreciate that an
appropriate buffer may be
chosen for inclusion in compositions of the invention based on its pKa and
other properties.
Buffers are well known in the art. Accordingly, the buffers described herein
are not intended
to constitute an exhaustive list, but are provided merely as exemplary buffers
that may be
used in the compositions of the invention. In certain aspects, a buffer may
include one or
more of the following: Tris, Tris HC I, potassium phosphate, sodium phosphate,
sodium
citrate, sodium ascorbate, combinations of sodium and potassium phosphate,
Tris/Tris HC 1,
sodium bicarbonate, arginine phosphate, arginine hydrochloride, histidine
hydrochloride,
cacodylate, succinate, 2-(N-morpholino)ethanesulfonic acid (MES), maleate, bis-
tris,
phosphate, carbonate, and any pharmaceutically acceptable salts and/or
combinations thereof
CA 03148275 2022-2-16

WO 2021/038078 36
PCT/EP2020/074134
In some aspects, it may be beneficial to include a surfactant in the
compositions of the
invention. Surfactants, in general, decrease the surface tension of a liquid
composition. This
may provide beneficial properties such as improved ease of filtration_
Surfactants also may act
as emulsifying agents and/or solubilizing agents. Surfactants are well known
in the art.
Accordingly, the surfactants described herein are not intended to constitute
an exhaustive list,
but are provided merely as exemplary surfactants that may be used in the
compositions of the
invention. Surfactants that may be included include, but are not limited to,
sorbitan esters
such as polysorbates (e.g., polysorbate 20 and polysorbate 80),
lipopolysaccharides,
io polyethylene glycols (e g , PEG 400 and PEG 3000), poloxamers (i.e.,
pluronics), ethylene
oxides and polyethylene oxides (e.g., Triton X-100), saponins, phospholipids
(e.g., lecithin),
and combinations thereof.
In some aspects, it may be beneficial to include a tonicity-adjusting agent in
the compositions
of the invention. The tonicity of a liquid composition is an important
consideration when
administering the composition to a patient, for example, by parenteral
administration
Tonicity-adjusting agents, thus, may be used to help make a formulation or
composition
suitable for administration. Tonicity-adjusting agents are well known in the
art Accordingly,
the tonicity-adjusting agents described herein are not intended to constitute
an exhaustive list,
but are provided merely as exemplary tonicity-adjusting agents that may be
used in the
compositions of the invention. Tonicity-adjusting agents may be ionic or non-
ionic and
include, but are not limited to, inorganic salts, amino acids, carbohydrates,
sugars, sugar
alcohols, and carbohydrates. Exemplary inorganic salts may include sodium
chloride,
potassium chloride, sodium sulfate, and potassium sulfate. An exemplary amino
acid is
glycine. Exemplary sugars may include sugar alcohols such as glycerol,
propylene glycol,
glucose, sucrose, lactose, and mannitol.
In some aspects, it may be beneficial to include a stabilizing agent in the
compositions of the
invention. Stabilizing agents help increase the stability of a therapeutically
effective substance
in compositions of the invention. This may occur by, for example, reducing
degradation or
preventing aggregation of a therapeutically effective substance. Without
wishing to be bound
by theory, mechanisms for enhancing stability may include sequestration of the

therapeutically effective substance from a solvent or inhibiting free radical
oxidation of the
anthracycline compound. Stabilizing agents are well known in the art
Accordingly, the
CA 03148275 2022-2-16

WO 2021/038078 37
PCT/EP2020/074134
stabilizing agents described herein are not intended to constitute an
exhaustive list, but are
provided merely as exemplary stabilizing agents that may be used in the
compositions of the
invention. Stabilizing agents may include, but are not limited to, emulsifiers
and surfactants.
The compositions as disclosed herein can be administered in a variety of
conventional ways.
In some aspects, the compositions of the invention are suitable for parenteral
administration
These compositions may be administered, for example, intraperitoneally,
intravenously,
intrarenally, intrathecally or by inhalation. In some aspects, the
compositions of the invention
are injected intravenously. One of skill in the art would appreciate that a
method of
io administering a therapeutically effective substance formulation or
composition of the
invention would depend on factors such as the age, weight, and physical
condition of the
patient being treated, and the disease or condition being treated. The skilled
worker would,
thus, be able to select a method of administration optimal for a patient on a
case-by-case
basis.
Angiotensin-receptor-agonist and/ or a precursor thereof can be administered
in any suitable
way, but are typically administered by continuous infusion. Accordingly,
increasing or
decreasing a rate of administration can be accomplished by changing the rate
of flow of an
intravenous drip, changing the concentration of the agent in an intravenous
drip, etc.
However, the manner in which the rate of administration is changed will depend
on the mode
of administration of the therapeutic. Where the therapeutic is administered
transmucosally or
transdemially, the rate may be increased by changing to a higher-release-rate
patch or
transdennal composition for example. Where the therapeutic is administered
orally, the rate
may be increased by switching to a higher-dose form, administering additional
doses, or
administering controlled-release dosage forms with a higher rate of release,
for example.
Where the therapeutic is administered by inhalation, the rate may be increased
by
administering additional boluses, a more concentrated bolus, or a faster-
release bolus, for
example. Other modes of administration (via subcutaneous injection pump,
suppository, etc.)
can be modulated in analogous fashions, and decreasing the rate of
administration can be
accomplished by doing the opposite of an action that would increase the rate
of administration
of the therapeutic.
CA 03148275 2022-2-16

WO 2021/038078 38
PCT/EP2020/074134
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
thereof for use in the treatment of shock in a subject that either runs into
shock or that has
developed shock, wherein said Angiotensin-Receptor-Agonist and/ or a precursor
thereof is
Angiotensin II that is administered at a rate between 0.1 and 200 ng/kg/min,
preferably
between 1 and 100 ng/kg/min, more preferred between 2 and 80 ng/kg/min, even
more
preferred between 5 and 60 ng/kg/min, even more preferred between 10 and 50
ng/kg/min,
even more preferred between 15 and 40 ng/kg/min most preferred at a rate of 20
ng/kg/min.
In a specific embodiment of the present disclosure the starting dosage
(initial rate) of
to angiotensin Ills 80 ng/kg/min, more preferred 40 ng/kg/min, most
preferred 20 ng/kg/min via
continuous intravenous infusion.
In another specific embodiment of the invention for titration of angiotensin
lithe blood
pressure response (e.g. mean arterial pressure; MAP) is monitored. Titration
of angiotensin II
may be carried out every 60 minutes, more preferred every 45 minutes, even
more preferred
every 30 minutes, even more preferred every 15 minutes, even more preferred
every 10
minutes, most preferred every 5 minutes. Angiotensin II titration may be
carried out by
increments of up to 40 ng/kg/min, more preferred up to 20 ng/kg/min, most
preferred up to
15 ng/kg/min as needed to achieve or maintain target blood pressure. In
another preferred
embodiment the dosage should not exceed 80 ng/kg/min of angiotensin II during
the first 3
hours of treatment. It is most preferred that maintenance doses should not
exceed
40 ng/kg/min andd oses as low as 1.25 ng/kg/min may be used.
In some embodiments, the patient has an initial mean arterial pressure (MAP)
of no more than
about 40 mm Hg, about 45 mm Hg, about 50 mm Hg, 55 mm Hg, about 60 mm Hg,
about
65 mm Hg, about 70 mm Hg, or about 75 mm Hg prior to administering the
composition. The
method may comprise measuring a mean atrial blood pressure of the patient and
increasing
the rate of administering angiotensin II if the mean arterial blood pressure
is less than about
40 mm Hg, about 45 mm Hg, about 50 mm Hg, 55 mm Hg, about 60 mm Hg, about
65 mm Hg, about 70 mm Hg, or about 75 mm Hg.
In one embodiment, the patient may be receiving a vasopressor (e.g., a
catecholamine, such as
norepinephtine, a norepinephrine equivalent, epinephrine, dopamine,
phenylephrine) or a
CA 03148275 2022-2-16

WO 2021/038078 39
PCT/EP2020/074134
combination thereof. In some embodiments, the vasopressor is vasopressin
(e.g., terlipressin,
argipressin, desmopressin, felypressin, lypressin, or ornipressin).
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
thereof for use in the treatment of shock in a subject that either runs into
shock or that has
developed shock, wherein said angiotensin-receptor-agonist and/ or a precursor
thereof, in
particular Angiotensin II, is administered in combination with an inhibitor of
DPP3.
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
io thereof for use in the treatment of shock in a subject that either runs
into shock or that has
developed shock, in combination with an inhibitor of DPP3, wherein said
inhibitor of DPP3 is
selected from the group comprising an anti-DPP3 antibody or anti-DPP3 antibody
fragment or
anti-DPP3 non-Ig scaffold.
In accordance with the invention the "anti-DPP3 antibody" is an antibody that
binds
specifically to DPP3, an "anti-DPP3 antibody fragment" is a fragment of said
anti-DPP3
antibody, wherein said fragment binds specifically to DPP3. An "anti-DPP3 non-
Ig scaffold"
is a non-Ig scaffold that binds specifically to DPP3.
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
thereof for use in the treatment of shock in a subject that either runs into
shock or that has
developed shock, in combination with an inhibitor of DPP3, wherein said
inhibitor of DPP3 is
an anti-DPP3 antibody or anti-DPP3 antibody fragment or anti-DPP3 non-Ig
scaffold that
binds to SEQ ID NO. 1, in particular that binds to SEQ ID NO. 2.
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
thereof for use in the treatment of shock in a subject that either runs into
shock or that has
developed shockõ in combination with an inhibitor of DPP3, wherein said
inhibitor of DPP3
is an antibody or fragment or scaffold that exhibits a minimum binding
affinity to DPP3 of
equal or less than 10-7 M.
In accordance with the present invention, the person skilled in the art is
well aware that the
binding affinity of the herein disclosed DPP3 binder to DPP3 may be measured
by various
CA 03148275 2022-2-16

WO 2021/038078 40
PCT/EP2020/074134
suitable assays known in the art. Respective examples are given below, but
these shall be not
construed as limiting possibilities to measure binding affinity of the herein
disclosed DPP3
binder to DPP3.
For example, the binding affinity of the DPP3 binder to an epitope may be
determined. A
binding assay may be performed to detect and/or quantitate antibody binding to
e.g. the
immunization peptide of the respective binder. For example, this immunization
peptide may
be immobilized upon a solid phase. The test sample (e.g. antibody solution) is
passed over the
immobile immunization peptide, and bound antibody can be detected. For the
purposes of the
present description, the term "solid phase" may be used to include any
material or vessel in
io which or on which the assay may be performed and includes, but
is not limited to, porous
materials, nonporous materials, test tubes, wells, slides, magnetic beads etc.
Exemplary detection methods:
- Label antibody before contacting with solid phase and detect respective
label
(fluorescence, chemiluminescence, enzymatic etc.).
- Use labeled secondary antibody against specific Fc part of sample-
antibody. Incubate
solid phase bound antibody with secondary antibody (e.g. anti human IgG, anti
murine
IgG) and detect respective label (fluorescence, chemiluminescence, enzymatic
etc.).
- Use a labeled antibody as competitor for solid phase binding (e.g.
labeled AlC1967).
- Quantify binding affinity by decrease of signal.
Another method to determine the affinity of antibodies to DPP3, the kinetics
of binding of
DPP3 to immobilized antibody may be determined by means of label-free surface
plasmon
resonance using a Biacore 2000 system (GE Healthcare Europe GmbH, Freiburg,
Germany).
Reversible immobilization of the antibodies may be performed using an anti-
mouse Fc
antibody covalently coupled in high density to a CM5 sensor surface (mouse
antibody capture
kit; GE Healthcare) (Lorenz et al. 2011. Antimicrob Agents Chemother. 55(1):
165-173)
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
thereof for use in the treatment of shock in a subject that either runs into
shock or that has
developed shock, in combination with an inhibitor of DPP3, wherein said
inhibitor of DPP3 is
CA 03148275 2022-2-16

WO 2021/038078 41
PCT/EP2020/074134
an antibody or fragment or scaffold that is monospecific, in one embodiment
said inhibitor of
DPP3 is an antibody or fragment or scaffold that is monoclonal.
Monospecific antibodies or fragments or non-Ig scaffolds according to the
invention are
antibodies or fragments or non-Ig scaffolds that all have affinity for the
same antigen.
Monoclonal antibodies are monospecific, but monospecific antibodies may also
be produced
by other means than producing them from a common germ cell.
In a specific embodiment said capture-binder that binds to frill-length DPP3
specifically
inhibits less than 50% DPP3 activity in a liquid phase assay, preferably less
than 40%, more
io preferably 30 %. For definition of liquid phase assay see above. In one
specific embodiment
to prevent inhibition of DPP3 the capture-binder should not bind DPP3 in the
area around the
active center and substrate binding region (amino acids 316 ¨ 669 of SEQ ID
No. 1).
One embodiment of the invention is an angiotensin-receptor-agonist and/ or a
precursor
thereof for use in the treatment of shock in a subject that either runs into
shock or that has
developed shock, in combination with an inhibitor of DPP3, wherein said
inhibitor of DPP3 is
an antibody or fragment or scaffold that binds to full-length DPP3 and
inhibits activity of
DPP3 of at least 10%, or at least 50%, more preferred at least 60%, even more
preferred more
than 70 %, even more preferred more than 80 %, even more preferred more than
90 %, even
more preferred more than 95 %.
Inhibition of DPP3 activity in a liquid phase assay by a binder may be
determined as
followed: Possible DPP3 capture-binders are incubated with recombinant or
purified native
DPP3 and specific DPP3 substrates in a liquid phase assay. Preferably, as
capture-binder for
the ECA is chosen the one with the least inhibitory ability. The capture-
binder should inhibit
DPP3 activity less than 50 %, preferably less than 40 %, preferably less than
30 A_ The
specific liquid phase DPP3 activity assay to determine the inhibitory ability
of possible
capture-binders comprises the following steps:
= Incubation of 25 ng/ ml native human DPP3 with 5 pg/ ml of the respective
capture-
binder and buffer control in 50 m.M Tris-HCI, pH 7,5 and 100 MM ZnC12 for 1
hour at
room temperature
= Addition of the fluorogenic substrate Arg-Arg-13NA (20 Ml, 2 mM).
CA 03148275 2022-2-16

WO 2021/038078 42
PCT/EP2020/074134
= Incubation at 37 C and monitoring the generation of free I3NA in a
Twinkle LB 970
microplate fluorometer (Berthold Technologies GmbH) over 1 hour. Fluorescence
of
I3NA is detected by exciting at 340 nm and measuring emission at 410 rim.
= Slopes (in RFU/ min) of increasing fluorescence of the different samples
are
calculated. The slope of native human DPP3 with buffer control is appointed as
100 %
activity. The inhibitory ability of a possible capture-binder is defined as
the decrease
of native human DPP3 activity by incubation with said capture-binder in
percent.
In a liquid phase assay samples of bodily fluids are directly subjected to
fluorogenic
substrates (e.g. Arg-Arg-13-NA). Since there are many different amino
peptidases in the
plasma (Sanden/1k et aL 1988), it is possible that the used substrate is
cleaved by peptidases
other than DPP3. To circumvent this problem one preferred method of detecting
specific
DPP3 activity is the use of an enzyme capture activity assay.
In one specific embodiment determination of active DPP3 in an enzyme capture
assay
comprises the steps.
= Contacting said sample with a capture-binder that binds to full-length
DPP3 but
preferably inhibits DPP3 activity in a liquid phase assay less than 50%,
preferably less
than 40%, more preferably 30 %. To prevent inhibition of DPP3 the capture-
binder
should not bind DPP3 in the area around the active center and substrate
binding region
(amino acids 316 ¨ 669 of SEQ ID No. 1),
= separating DPP3 bound to said capture binder from bodily fluid sample,
= adding substrate of DPP3 to said separated DPP3,
= quantifying DPP3 activity by measuring the conversion of the substrate of
DPP3.
An "antibody" according to the present invention is a protein including one or
more
polypeptides substantially encoded by immunoglobulin genes that specifically
binds an
antigen. The recognized immunoglobulin genes include the kappa, lambda, alpha
(IgA),
gamma (IgGI, IgG2, IgG3, IgG4), delta (IgD), epsilon (IgE) and mu (104)
constant region
genes, as well as the myriad immunoglobulin variable region genes. Full-length

immunoglobulin light chains are generally about 25 lcDa or 214 amino acids in
length.
CA 03148275 2022-2-16

WO 2021/038078 43
PCT/EP2020/074134
Full-length immunoglobulin heavy chains are generally about 50 kDa or 446
amino acids in
length. Light chains are encoded by a variable region gene at the NH2-terminus
(about 110
amino acids in length) and a kappa or lambda constant region gene at the COOH-
terminus.
Heavy chains are similarly encoded by a variable region gene (about 116 amino
acids in
length) and one of the other constant region genes.
The basic structural unit of an antibody is generally a tetramer that consists
of two identical
pairs of immunoglobulin chains, each pair having one light and one heavy
chain. In each pair,
the light and heavy chain variable regions bind to an antigen, and the
constant regions mediate
io effector functions. Immunoglobulins also exist in a variety of other forms
including, for
example, Fv, Fab, and F(a131)2, as well as bifunctional hybrid antibodies and
single chains
(e.g., Lanzavecchia et aL, Fur. .1 linmutiol. 17:105,1987; Huston et al.,
Proc. Natl. Acad Sc!.
U.S.A., 85:5879-5883, 1988; Bird et aL, Science 242:423-426, 1988; Hood et aL,

Immunology. Benjamin. N.Y. 2nd ed.. 1984; Hunkapiller and Hood. Nature 323:15-
16.1986).
An immunoglobulin light or heavy chain variable region includes a framework
region
interrupted by three hypervariable regions, also called complementarity
determining regions
(CDR's) (see, Sequences of Proteins of Immunological Interest, E. Kabat et aL,
U.S.
Department of Health and Human Services, 1983). As noted above, the CDRs are
primarily
responsible for binding to an epitope of an antigen. An immune complex is an
antibody, such
as a monoclonal antibody, chimeric antibody, humanized antibody or human
antibody, or
functional antibody fragment, specifically bound to the antigen.
"Chimeric antibodies" are antibodies whose light and heavy chain genes have
been
constructed, typically by genetic engineering, from immunoglobulin variable
and constant
region genes belonging to different species. For example, the variable
segments of the genes
from a mouse monoclonal antibody can be joined to human constant segments,
such as kappa
and gamma 1 or gamma 3. In one example, a therapeutic chimeric antibody is
thus a hybrid
protein composed of the variable or antigen-binding domain from a mouse
antibody and the
constant or effector domain from a human antibody, although other mammalian
species can
be used, or the variable region can be produced by molecular techniques.
Methods of making
chimeric antibodies are well known in the art, e.g., see U.S. Patent No.
5,807,715. A
"humanized" immunoglobulin is an immunoglobulin including a human framework
region
and one or more CDRs from a non-human (such as a mouse, rat, or synthetic)
CA 03148275 2022-2-16

WO 2021/038078 44
PCT/EP2020/074134
immunoglobulin. The non-human immunoglobulin providing the CDRs is termed a
"donor"
and the human immunoglobulin providing the framework is termed an "acceptor."
In one embodiment of the invention, all the CDRs are from the donor
immunoglobulin in a
humanized immunoglobulin. Constant regions need not be present, but if they
are, they must
be substantially identical to human immunoglobulin constant regions, i.e., at
least about
85-90%, such as about 95% or more identical. Hence, all parts of a humanized
immunoglobulin, except possibly the CDRs, are substantially identical to
corresponding parts
of natural human immunoglobulin sequences.
io A "humanized antibody" in accordance with the invention is an antibody
comprising a
humanized light chain and a humanized heavy chain immunoglobulin. A humanized
antibody
binds to the same antigen as the donor antibody that provides the CDRs. The
acceptor
framework of a humanized immunoglobulin or antibody may have a limited number
of
substitutions by amino acids taken from the donor framework. Humanized or
other
monoclonal antibodies can have additional conservative amino acid
substitutions which have
substantially no effect on antigen binding or other immunoglobulin functions.
Exemplary
conservative substitutions are those such as gly, ala; val, ile, leu; asp,
glu; asn, gin; ser, thr;
lys, arg; and phe, tyr. Humanized immunoglobulins can be constructed by means
of genetic
engineering (e.g., see U.S. Patent No. 5,585,089). A human antibody is an
antibody wherein
the light and heavy chain genes are of human origin. Human antibodies can be
generated
using methods known in the art. Human antibodies can be produced by
immortalizing a
human B cell secreting the antibody of interest. Immortalization can be
accomplished, for
example, by EBV infection or by fusing a human B cell with a myeloma or
hybridoma cell to
produce a trioma cell. Human antibodies can also be produced by phage display
methods (see,
e.g., Dower et PCT Publication No. WO 91/17271; McCafferty et al., PCT
Publication
No. WO 92/001047; and Winter, PCT Publication No. WO 92/20791), or selected
from a
human combinatorial monoclonal antibody library (see the Morphosys website).
Human
antibodies can also be prepared by using transgenic animals carrying a human
immunoglobulin gene (for example, see Lonberg et al., PCT Publication No WO
93/12227;
and Kucherlapati, PCT Publication No. WO 91/10741).
Thus, the anti-DPP3 antibody or anti-DPP3 antibody fragment in accordance with
the
invention may have the formats known in the art. Examples are human
antibodies,
CA 03148275 2022-2-16

WO 2021/038078 45
PCT/EP2020/074134
monoclonal antibodies, humanized antibodies, chimeric antibodies, CDR-grafted
antibodies
or antibody fragments thereof, but not limited to.
In a specific embodiment of the invention the anti-DPP3 antibody is a
monoclonal antibody or
a fragment thereof In one embodiment of the invention the anti-DPP3 antibody
or the
anti-DPP3 antibody fragment is a human or humanized antibody or derived
therefrom. In one
specific embodiment one or more (murine) CDR's are grafted into a human
antibody or
antibody fragment.
io In a preferred embodiment antibodies according to the present invention are
recombinantly
produced antibodies as e.g. IgG, a typical full-length immunoglobulin, or
antibody fragments
containing at least the F-variable domain of heavy and/or light chain as e.g.
chemically
coupled antibodies (fragment antigen binding) including but not limited to Fab-
fragments
including Fab minibodies, single chain Fab antibody, monovalent Fab antibody
with epitope
tags, e.g. Fab-V5Sx2; bivalent Fab (mini-antibody) dimerized with the CH3
domain; bivalent
Fab or multivalent Fab, e.g. formed via multimerization with the aid of a
heterologous
domain, e.g. via dimerization of dHLX domains, e.g. Fab-dHLX-FSx2; F(ab`)2-
fragments,
scFv-fragments, multimerized multivalent and/or multispecific scFv-fragments,
bivalent
and/or bispecific diabodies, BITE (bispecific T-cell engager), ttifunctional
antibodies,
polyvalent antibodies, e.g. from a different class than G; single-domain
antibodies, e.g.
nanobodies derived from camelid or fish immunoglobulines and numerous others.
In addition to anti-DPP3 antibodies or anti-DPP3 antibody fragments, other
biopolymer
scaffolds, so called non-Ig scaffolds, are well known in the art to complex a
target molecule
and have been used for the generation of highly target specific biopolymers.
Examples are
aptamers, spiegelmers, anticalins and conotoxins.
Non-Ig scaffolds with the context of the invention may be protein scaffolds
and may be used
as antibody mimics as they are capable to bind to ligands or antigens. Non-Ig
scaffolds may
be selected from the group comprising tetranectin-based non-Ig scaffolds (e.g.
described in
US 2010/0028995), fibronectin scaffolds (e.g. described in EP 1266 025;
lipocalin-based
scaffolds (e.g. described in WO 2011/154420); ubiquitin scaffolds (e.g.
described in
WO 2011/073214), transferring scaffolds (e.g. described in US 2004/0023334),
protein A
scaffolds (e.g. described in EP 2231860), ankyrin repeat based scaffolds (e.g.
described in
CA 03148275 2022-2-16

WO 2021/038078 46
PCT/EP2020/074134
WO 2010/060748), microprotein (preferably microproteins forming a cystine
knot) scaffolds
(e.g. described in EP 2314308), Fyn SH3 domain based scaffolds (e.g. described
in
WO 2011/023685), EGFR-A-domain based scaffolds (e.g. described in WO
2005/040229)
and Kunitz domain based scaffolds (e.g. described in EP 1941867). Non-Ig
scaffolds may be
peptide or oligonucleotide aptamers. Aptamers are usually created by selecting
them from a
large random sequence pool and are either short strands of oligonucleotides
(DNA, RNA or
XNA; Xu et ad. 2010. Deng et al. 2014) or short variable peptide domains
attached to a
protein scaffold (Li et aL 2011).
m In an alternative embodiment the anti-DPP3 antibody format is selected from
the group
comprising Fv fragment, scFy fragment, Fab fragment, scFab fragment, F(ab)2
fragment and
scFv-Fc Fusion protein. In another preferred embodiment the antibody format is
selected from
the group comprising scFab fragment, Fab fragment, scFv fragment and
bioavailability
optimized conjugates thereof, such as PEGylated fragments.
With the context of the invention, the term "antibody" generally comprises
monoclonal and
polyclonal antibodies and binding fragments thereof, in particular Fc-
fragments as well as so
called "single-chain-antibodies" (Bird et aL 1988), chimeric, humanized, in
particular
CDR-grafted antibodies, and di- or tetrabodies (Holliger et aL 1993). Also
comprised are
immunoglobulin-like proteins that are selected through techniques including,
for example,
phage display to specifically bind to the molecule of interest contained in a
sample. In this
context the term "specific binding" refers to antibodies raised against the
molecule of interest
or a fragment thereof. An antibody is considered to be specific, if its
affinity towards the
molecule of interest or the aforementioned fragment thereof is at least
preferably 50-fold
higher, more preferably 100-fold higher, most preferably at least 1000-fold
higher than
towards other molecules comprised in a sample containing the molecule of
interest. It is well
known in the art how to make antibodies and to select antibodies with a given
specificity.
In a specific embodiment of the invention said anti-DPP3 antibody or anti-DPP3
antibody
fragment binding to an epitope according to SEQ ID NO.: 2, wherein said
epitope is
comprised in a DPP3 protein or functional derivative thereof is a monoclonal
antibody or a
monoclonal antibody fragment thereof In one embodiment of the invention the
anti-DPP3
antibody or the anti-DPP3 antibody fragment binding to an epitope according to
SEQ ID NO..
2, wherein said epitope is comprised in a DPP3 protein or functional
derivative thereof is a
CA 03148275 2022-2-16

WO 2021/038078 47
PCT/EP2020/074134
human or humanized antibody or derived therefrom or humanized antibody
fragment or
derived therefrom.
In one specific embodiment one or more (murine) CDR's are grafted into a human
antibody
or antibody fragment.
In another aspect of the invention, the provided subject matter is a human CDR-
grafted anti-
DPP3 antibody or anti-DPP3 antibody fragment thereof that is directed to and
binding to an
epitope according to SEQ ID NO.: 2, wherein said epitope is comprised in a
DPP3 protein or
io a functional derivative thereof, and wherein said human CDR-grafted anti-
DPP3 antibody or
anti-DPP3 antibody fragment thereof comprises an antibody heavy chain variable
region
(H chain) comprising:
SEQ ID NO.: 4
and/or further comprises an antibody light chain variable region (L chain)
comprising:
SEQ ID NO.: 5.
Further subject matter of the present invention in another aspect is a human
CDR-grafted anti-
DPP3 antibody or anti-DPP3 antibody fragment thereof that is directed to and
binding to an
epitope according to SEQ ID NO.: 2, wherein said epitope is comprised in a
DPP3 protein or
a functional derivative thereof, and wherein the said human CDR-grafted anti-
DPP3 antibody
or anti-DPP3 antibody fragment thereof comprises an antibody heavy chain
variable region
(I-1 chain) comprising:
SEQ ID NO.: 11
and/or further comprises an antibody light chain variable region (L chain)
comprising:
SEQ ID NO.: 12.
In one specific embodiment of the invention subject matter of the present
invention is a
human monoclonal anti-DPP3 antibody or monoclonal anti-DPP3 antibody fragment
thereof
that is directed to and binding to an epitope according to SEQ ID NO.: 2,
wherein said epitope
is comprised in a DPP3 protein or a functional derivative thereof, and wherein
the heavy
chain comprises at least one CDR of:
SEQ ID NO.: 6, SEQ ID NO.: 7 or SEQ ID NO.: 8
and wherein the light chain comprises at least one CDR of:
SEQ ID NO.: 9, KVS or SEQ ID NO.: 10.
CA 03148275 2022-2-16

WO 2021/038078 48
PCT/EP2020/074134
The amount of DPP3 protein and/ or DPP3 activity in a sample of bodily fluid
of said subject
may be determined by different methods, e.g. immunoassays, acitivity assays,
mass
spectrometric methods etc.
The amount of DPP3 protein and/ or DPP3 activity in a sample of bodily fluid
of said subject
may be determined for example by one of the following methods:
1. Luminescence immunoassay for the quantification of DPP3 protein
concentrations
(LIA) (Rehfeld et al. 2019 ALM 3(6):943-953).
The LIA is a one-step chemiluminescence sandwich immunoassay that uses white
high-
binding polystyrene microtiter plates as solid phase. These plates are coated
with monoclonal
anti-DPP3 antibody AK2555 (capture antibody). The tracer anti-DPP3 antibody
AK2553 is
labeled with MA70-acridinium-NHS-ester and used at a concentration of 20 ng
per well
Twenty microliters of samples (e.g. serum, heparin-plasma, citrate-plasma or
EDTA-plasma
derived from patients' blood) and calibrators are pipetted into coated white
microtiter plates.
After adding the tracer antibody AK2553, the micro-titer plates are incubated
for 3 h at room
temperature and 600 rpm. Unbound tracer is then removed by 4 washing steps
(350 itL per
well). Remaining chemiluminescence is measured for is per well by using a
microtiter plate
luminometer. The concentration of DPP3 is determined with a 6-point
calibration curve.
Calibrators and samples are preferably run in duplicate.
2. Enzyme capture activity assay for the quantification of DPP3 activity (ECA)
(Rehfeld
et aL 2019 JALlit 3(6):943-953).
The ECA is a DPP3-specific activity assay that uses black high-binding
polystyrene microtiter
plates as solid phase. These plates are coated with monoclonal anti-DPP3
antibody AK2555
(capture antibody). Twenty microliters of samples (e.g. serum, heparin-plasma,

citrate-plasma, EDTA-plasma, cerebrospinal fluid and urine) and calibrators
are pipetted into
coated black microtiter plates. After adding assay buffer (200 gL), the
microtiter plates are
incubated for 2 h at 22 C and 600 rpm. DPP3 present in the samples is
immobilized by
binding to the capture antibody. Unbound sample components are removed by 4
washing
steps (350 pL per well). The specific activity of immobilized DPP3 is measured
by the
CA 03148275 2022-2-16

WO 2021/038078 49
PCT/EP2020/074134
addition of the fluorogenic substrate, Arg-Arg-13-Naphthylamide (Arg2-I3NA),
in reaction
buffer followed by incubation at 37 C for 1 h. DPP3 specifically cleaves Arg2-
I3NA into
Arg-Arg dipeptide and fluorescent fl-naphthylamine. Fluorescence is measured
with a
fluorometer using an excitation wavelength of 340 nm and emission is detected
at 410 nm.
The activity of DPP3 is determined with a 6-point calibration curve.
Calibrators and samples
are preferably run in duplicates.
3. Liquid-phase assay for the quantification of DPP3 activity (LAA) (modified
from
Jones et al., Analytical Biochemistry. 1982).
The LAA is a liquid phase assay that uses black non-binding polystyrene
microliter plates to
measure DPP3 activity. Twenty microliter of samples (e.g. serum, heparin-
plasma, citrate-
plasma) and calibrators are pipetted into non-binding black microtiter plates.
After addition of
fluorogenic substrate, Arg2-)NA, in assay buffer (200 piL), the initial I3NA
fluorescence
(T=0) is measured in a fluorimeter using an excitation wavelength of 340 nm
and emission is
detected at 410 nm. The plate is then incubated at 37 C for 1 hour. The final
fluorescence of
(T=60) is measured. The difference between final and initial fluorescence is
calculated. The
activity of DPP3 is determined with a 6-point calibration curve. Calibrators
and samples are
preferably run in duplicates.
A variety of immunoassays are known and may be used for the assays and methods
of the
present invention, these include: radioimmunoassays ("MA") homogeneous enzyme-
multiplied immunoassays ("EMIT"), enzyme linked immunoadsorbent assays
("ELISA"),
apoenzyme reactivation immunoassay ("ARIS"), chemiluminescence- and
fluorescence-
immunoassays, Luminex-based bead arrays, protein microarray assays, and rapid
test formats
such as for instance immunochromatographic strip tests ("dipstick
immunoassays") and
immuno-chromotography assays.
In one embodiment of the invention such an assay is a sandwich immunoassay
using any kind
of detection technology including but not restricted to enzyme label,
chemiluminescence
label, electrochemiluminescence label, preferably a fully automated assay. In
one embodiment
of the invention such an assay is an enzyme labeled sandwich assay. Examples
of automated
or fully automated assay comprise assays that may be used for one of the
following systems:
CA 03148275 2022-2-16

WO 2021/038078 50
PCT/EP2020/074134
Roche Elecsys , Abbott Architect , Siemens Centauer , Brahms 1Cryptor ,
Biomerieux
Vidas , Al ere Triage .
In one embodiment of the invention it may be a so-called POC-test (point-of-
care), that is a
test technology which allows performing the test within less than 1 hour near
the patient
without the requirement of a fully automated assay system. One example for
this technology
is the immunochromatographic test technology.
In a specific embodiment of the invention said POC-test combines the
measurement of more
than one analyte at the same time. In another specific embodiment of the
invention said
analyte is selected from the group of DPP3 and Pro-adrenomedullin or fragments
thereof In a
io very specific embodiment of the invention said POC-test combines the
measurement of DPP3
and mature ADM.
In one embodiment of the invention at least one of the binders is labeled in
order to be
detected.
In a preferred embodiment said label is selected from the group comprising
chemiluminescent
label, enzyme label, fluorescence label, radioiodine label.
The assays can be homogenous or heterogeneous assays, competitive and non-
competitive
assays. In one embodiment, the assay is in the form of a sandwich assay, which
is a
non-competitive immunoassay, wherein the molecule to be detected and/or
quantified is
bound to a first antibody and to a second antibody_ The first antibody may be
bound to a solid
phase, e.g. a bead, a surface of a well or other container, a chip or a strip,
and the second
antibody is an antibody which is labeled, e.g. with a dye, with a
radioisotope, or a reactive or
catalytically active moiety. The amount of labeled antibody bound to the
analyte is then
measured by an appropriate method. The general composition and procedures
involved with
"sandwich assays" are well-established and known to the skilled person (The
Immunoassay
Handbook, Ed David Wilcl Elsevier LTD, Oxford; 3rd ed. (May 2005), ISBN-11:
978-
0080445267; Hultschig C et al., Curr Opin Chem BioL 2006 Feb;10(1):440. PligD:

16376134).
In another embodiment the assay comprises two capture molecules, preferably
antibodies
which are both present as dispersions in a liquid reaction mixture, wherein a
first labelling
component is attached to the first capture molecule, wherein said first
labelling component is
part of a labelling system based on fluorescence- or chemiluminescence-
quenching or
amplification, and a second labelling component of said marking system is
attached to the
CA 03148275 2022-2-16

WO 2021/038078 51
PCT/EP2020/074134
second capture molecule, so that upon binding of both capture molecules to the
analyte a
measurable signal is generated that allows for the detection of the formed
sandwich
complexes in the solution comprising the sample.
In another embodiment, said labeling system comprises rare earth cryptates or
rare earth
chelates in combination with fluorescence dye or chemiluminescence dye, in
particular a dye
of the cyanine type.
In the context of the present invention, fluorescence based assays comprise
the use of dyes,
which may for instance be selected from the group comprising FAM (5-or
6-carboxyfluorescein), VIC, NED, Fluorescein, Fluoresceinisothiocyanate
(FITC),
io IRD-700/800, Cyanine dyes, auch as CY3, CY5, CY.3.5, CY5.5, Cy7,
Xanthen, 6-Carboxy-
2',4',7',4,7-hexachlorofluorescein (HEX),
TET, 6-Carboxy-4"5 -dichloro-2'
,7' -
dimethodyfluorescein (JOE), N,N,N',N'-Tetramethy1-6-carboxyrhodamine (TAMPA),
6-Carboxy-X-rhodamine (ROX), 5-Carboxyrhodamine-6G (R6G5), 6-carboxyrhodamine-
6G
(RG6), Rhodamine, Rhodamine Green, Rhodamine Red, Rhodamine 110, BODIPY dyes,
such as BOD1PY TMR, Oregon Green, Coumarines such as Umbelliferone,
Benzimides, such
as Hoechst 33258; Phenanthridines, such as Texas Red, Yakima Yellow, Alexa
Fluor, PET,
Ethidiumbromide, Acridinium dyes, Carbazol dyes, Phenoxazine dyes, Porphyrine
dyes,
Polymethin dyes, and the like.
In the context of the present invention, chemiluminescence based assays
comprise the use of
dyes, based on the physical principles described for chemiluminescent
materials in (Kirk-
Othmer. Encyclopedia of chemical technology. 4th ed., executive editor, J. I.
Kroschwitz;
editor. M. Howe-Grant, John Wiley & Sons, 1993, vol. 15+ p. 518-562,
incorporated herein by
reference, including citations on pages 551-562). Preferred chemiluminescent
dyes are
acridiniumesters.
As mentioned herein, an "assay" or "diagnostic assay" can be of any type
applied in the field
of diagnostics. Such an assay may be based on the binding of an analyte to be
detected to one
or more capture probes with a certain affinity. Concerning the interaction
between capture
molecules and target molecules or molecules of interest, the affinity constant
is preferably
greater than 108 M-1.
DPP3 activity can be measured by detection of cleavage products of DPP3
specific substrates.
Known peptide hormone substrates include Leu-enkephalin, Met-enkephalin,
endomorphin 1
and 2, valoiphin, I3-casomorphin, dynorphin, proctolin, ACTH
(Adrenocorticotropic
CA 03148275 2022-2-16

WO 2021/038078 52
PCT/EP2020/074134
hormone) and MSH (melanocyte-stimulating hormone; Abramie et al. 2000. Bar:sun
n at
2007. Dhanda n al. 2008). The cleavage of mentioned peptide hormones as well
as other
untagged oligopeptides (e.g. Ma-Ma-Ma-Ala, Dhandct et at 2008) can be
monitored by
detection of the respective cleavage products. Detection methods include, but
are not limited
to, HPLC analysis (e.g. Lee & Snyder 1982), mass spectrometry (e.g. Abrconie a
aL 2000),
HI-NMR analysis (e.g. Vandenberg et at 1985), capillary zone electrophoresis
(CE; e.g
flargun n at 2007), thin layer chromatography (e.g. Dhanda n al. 2008) or
reversed phase
chromatography (e.g. Iiilazocco et al. 2006).
io Detection of fluorescence due to hydrolysis of fluorogenic substrates by
DPP3 is a standard
procedure to monitor DPP3 activity. Those substrates are specific di- or
tripeptides (Arg-Arg,
Ala-Ala, Ala-Arg, Ala-Phe, Asp-Arg, Gly-Ma, Gly-Arg, Gly-Phe, Leu-Ala, Leu-
Gly, Lys-
Ala, Phe-Arg, Suc-Ma-Ma-Phe) coupled to a fluorophore. Fluorophores include
but are not
limited to I3-naphtylamide (2-naphtylamide, I3NA, 2NA), 4-methoxy-13-
naphtylamide
(4-methoxy-2-naphtylamide) and 7-amido-4-methylcoumarin (AMC, MCA; Abrarnit et
aL
2000. Ohkubo et at 1999). Cleavage of these fluorogenic substrates leads to
the release of
fluorescent P-naphtylamine or 7-amino-4-methylcoumarin respectively. In a
liquid phase
assay or an ECA substrate and DPP3 are incubated in for example a 96 well
plate format and
fluorescence is measured using a fluorescence detector (Ellis & Nuenke 1967).
Additionally,
DPP3 carrying samples can be immobilized and divided on a gel by
electrophoresis, gels
stained with fluorogenic substrate (e.g. Arg-Arg-PNA) and Fast Garnet GBC and
fluorescent
protein bands detected by a fluorescence reader (Ohkubo et at 1999). The same
peptides
(Arg-Arg, Ma-Ala, Ma-Mg, Ma-Phe, Asp-Mg, Gly-Ma, Gly-Arg, Gly-Phe, Leu-Ala,
Leu-Gly, Lys-Ma, Phe-Arg, Suc-Ma-Ma-Phe) can be coupled to chromophores, such
as
p-nitroanilide diacetate. Detection of color change due to hydrolysis of
chromogenic
substrates can be used to monitor DPP3 activity.
Another option for the detection of DPP3 activity is a Protease-GloThi Assay
(commercially
available at Promega). In this embodiment of said method DPP3 specific di- or
tripeptides
(Arg-Arg, Ma-Ala, Ala-Arg, Ma-Phe, Asp-Mg, Gly-Ala, Gly-Arg, (ily-Phe, Leu-
Ala,
Leu-Gly, Lys-Ma, Phe-Arg, Suc-Ma-Ma-Phe) are coupled to aminoluciferin. Upon
cleavage
by DPP3, aminoluciferin is released and serves as a substrate for a coupled
luciferase reaction
that emits detectable luminescence.
CA 03148275 2022-2-16

WO 2021/038078 53
PCT/EP2020/074134
In a preferred embodiment DPP3 activity is measured by addition of the
fluorogenic substrate
Arg-Arg-f3NA and monitoring fluorescence in real time.
In a specific embodiment of said method for determining active DPP3 in a
bodily fluid
sample of a subject said capture binder reactive with DPP3 is immobilized on a
solid phase.
The test sample is passed over the immobile binder, and DPP3, if present,
binds to the binder
and is itself immobilized for detection. A substrate may then be added, and
the reaction
product may be detected to indicate the presence or amount of DPP3 in the test
sample. For
io the purposes of the present description, the term "solid phase" may be
used to include any
material or vessel in which or on which the assay may be performed and
includes, but is not
limited to: porous materials, nonporous materials, test tubes, wells, slides,
agarose resins
(e.g. Sepharose from GE Healthcare Life Sciences), magnetic particals (e.g.
DynabeadsTm or
Piercer magnetic beads from Thermo Fisher Scientific), etc.
Binders of protein or peptide origin (e.g. antibody, antibody fragments, non-
Ig scaffold) are
immobilized onto the solid phase by methods comprising: physical adsorption
(e.g. by
electrostatic interaction or hydrophobic interaction), bioaffinity
immobilization (e.g. avidin-
biotin, protein Al G/ L, His-tag and Ni2+-NTA, GST-tag and gluthatione, DNA
hybridization,
aptamers), covalent bond (e.g. amine and N-hydroxysuccinimide) or a
combination of said
immobilization methods (Kim 41 Herr 2013). Binders of ol igonucleoti de origin

(e.g. aptamers) may be immobilized onto the solid phase by utilization of the
(strept)ayidin-
biotin system (Muller et al. 2012. Deng et at 2014).
In a specific embodiment of said method for determining DPP3 activity in a
bodily fluid
sample of a subject said separation step is a washing step that removes
ingredients of the
sample that are not bound to said capture-binder from the captured DPP3. That
separation
step can be any other step that separates DPP3 bound to said capture-binder
from the
ingredients of said bodily fluid sample.
In a specific embodiment of said method for determining DPP3 activity in a
bodily fluid
sample of a subject said DPP3 substrate conversion by immobilized DPP3 is
measured
(detected) by a method selected from the group comprising: fluorescence of
fluorogenic
substrates (e.g. Arg-Arg-I3NA, Arg-Arg-AMC), color change of chromogenic
substrates,
CA 03148275 2022-2-16

WO 2021/038078 54
PCT/EP2020/074134
luminescence of substrates coupled to aminoluciferin (Promega Protease-Glom
Assay), mass
spectrometry, HPLC/ FPLC (reversed phase chromatography, size exclusion
chromatography), thin layer chromatography, capillary zone electrophoresis,
gel
electrophoresis followed by activity staining (immobilized, active DPP3) or
western blot
(cleavage products).
In a specific embodiment of said method for determining DPP3 acitivity in a
bodily fluid
sample of a subject said substrate may be selected from the group comprising:
Leu-enkephalin, Met-enkephalin, endomorphin 1 and 2, valorphin, I3-
casomorphin,
io dynorphin, proctolin, ACTH and MSH, or di- and tri-peptides coupled to a
fluorophore, a
chromophore or aminoluciferin (Promega Protease-Glom Assay). Di- or
tripeptides that are
cleaved by DPP3 include, but are not limited to, Arg-Arg, Ala-Ala, Ala-Mg, Ala-
Phe,
Asp-Arg, Gly-Ala, Gly-Arg, Gly-Phe, Leu-Ala, Leu-Gly, Lys-Ala, Phe-Arg, Suc-
Ala-Ala-
Phe. Fluorophores include but are not limited to I3-naphtylamide (2-
naphtylamide, I3NA,
2NA), 4-methoxy-13-naphtylamide (4-methoxy-2-naphtylamide) and 7-amido-4-
methylcoumarin (AMC, MCA; Abramic et al. 2000, Ohkubo et at. 1999). Cleavage
of these
fluorogenic substrates leads to the release of fluorescent I3-naphtylamine or
7-amino-4-
methylcoumarin respectively. Chromophores include but are not limited to p-
nitroanilide
diacetate (pNA). The hydrolysis of a peptide-pNA bond in the chromogenic
substrates results
in the release of pNA which in turn changes color. Thus, the change in
absorbance (DA/min)
is directly proportional to the enzymatic activity. Using the Protease-Glom
Assay from
Promega, upon cleavage by DPP3, aminoluciferin is released and serves as a
substrate for a
coupled luciferase reaction that emits detectable luminescence.
Subject-matter of the present invention is also a method for prognosing an
outcome and/or the
risk of an adverse event in a subject that has developed refractory shock,
wherein said method
is comprising the steps:
= determining the level of DPP3 in a sample of bodily fluid of said
subject;
= comparing said level of determined DPP3 to a predetermined threshold,
= correlating said level of DPP3 with said risk of an adverse event in said
subject,
wherein an elevated level above a certain threshold is predictive for an
enhanced risk
of said adverse events or,
CA 03148275 2022-2-16

WO 2021/038078 55
PCT/EP2020/074134
= correlating said level of DPP3 with success of a therapy or intervention
in said subject,
wherein a level below a certain threshold is predictive for a success of
therapy or
intervention
In the context of the present invention, the term "prognosis" denotes a
prediction of how a
patient's medical condition will progress. This may include an estimation of
the chance of
recovery or the chance of an adverse event for said patient. Adverse event is
defined as organ
dysfunction or mortality. Organ dysfunction is defined as renal decline,
cardiac dysfunction or
liver dysfunction
ici Further embodiments of the present invention are:
1. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock, wherein said method is comprising the
steps:
= determining the level of DPP3 in a sample of bodily fluid of said subject;
= comparing said level of determined DPP3 to a predetermined threshold,
wherein said subject is predicted to run into refractory shock or is diagnosed
as having
refractory shock if said determined level of DPP3 is above said predetermined
threshold.
2. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiment 1, wherein said

shock is selected from the group comprising shock due to hypovolemia,
cardiogenic
shock, obstructive shock and distributive shock, in particular cardiogenic
shock or
septic shock.
3. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiment 1 and 2,
wherein
= in case of cardiogenic shock said subject may have suffered an acute
coronary
syndrome (e.g. acute myocardial infarction) or wherein said subject has heart
failure (e.g. acute decompensated heart failure), myocarditis, arrhythmia,
cardiomyopathy, valvular heart disease, aortic dissection with acute aortic
stenosis, traumatic chordal rupture or massive pulmonary embolism or
CA 03148275 2022-2-16

WO 2021/038078 56
PCT/EP2020/074134
= in case of hypovolemic shock said subject may have suffered a hemorrhagic

disease including gastrointestinal bleed, trauma, vascular etiologies (e.g.
ruptured abdominal aortic aneurysm, tumor eroding into a major blood vessel)
and spontaneous bleeding in the setting of anticoagulant use or a non-
hemorrhagic disease including vomiting, diarrhea, renal loss, skin
losses/insensible losses (e.g. bums, heat stroke) or third-space loss in the
setting of pancreatitis, cirrhosis, intestinal obstruction, trauma. or
= in case of obstructive shock said patient may have suffered a cardiac
tamponade, tension pneumothorax, pulmonary embolism or aortic stenosis, or
io
= in case of distributive shock said
patient may have septic shock, neurogenic
shock, anaphylactic shock or shock due to adrenal crisis.
4. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiments 1 to 3,
wherein said
method is used for initiation and/or termination and/ or stratification and/or
guidance
of treatment.
5. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to 4,
wherein a treatment is initiated and/or maintained and/or withheld and/ or
terminated
if said determined level of DPP3 is above said predetermined threshold.
6. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiment 5, wherein said
treatment is selected from the group of vasopressors, Angiotensin-Receptor-
Agonists
and/or precursors thereof, inhibitors of the DPP3 activity and anti-
adrenomedullin
antibodies or anti-adrenomedullin antibody fragments.
7. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to 6,
wherein either the level of DPP3 protein and/or the level of active DPP3 is
determined
and compared to a predetermined threshold.
CA 03148275 2022-2-16

WO 2021/038078 57
PCT/EP2020/074134
8. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to 7,

wherein the level of DPP3 is determined by contacting said sample of bodily
fluid
with a capture binder that binds specifically to DPP3.
9. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiment 8, wherein said

capture binder for determining the level of DPP3 may be selected from the
group of
antibody, antibody fragment or non-IgG scaffold.
io
10. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiments 8 to 9,
wherein said
capture binder is an antibody.
11. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 8 to
10,
wherein said capture binder is a monoclonal antibody.
12. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to
11,
wherein said sample of bodily fluid is selected from the group of whole blood,
plasma,
and serum.
13. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to
12,
wherein said method of diagnosing or predicting is conducted at least twice.
14. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to
13,
wherein a treatment with Angiotensin-Receptor-Agonists and/ or precursors
thereof
and/or inhibitors of the DPP3 activity is initiated and/or continued when the
level of
DPP3 in said sample is above a certain threshold and/or wherein a treatment
with
vasopressors is withheld and/ or terminated if said determined level of DPP3
is above
said predetermined threshold.
CA 03148275 2022-2-16

WO 2021/038078 58
PCT/EP2020/074134
15. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to
13,
wherein a treatment with vasopressors is initiated and/or continued when the
level of
DPP3 in said sample is below a certain threshold and/or wherein a treatment
with
Angiotensin-Receptor-Agonists and/ or precursors thereof and/or inhibitors of
the
DPP3 activity is withheld and/ or terminated if the said determined level of
DPP3 is
below said predetermined threshold.
lo
16. A method for predicting or
diagnosing a refractory shock in a subject that either runs
into shock or that has developed shock according to embodiments 14 to 15,
wherein in
addition the level of Pro-adrenomedullin or fragments thereof is determined
and
wherein treatment with an anti-ADM antibody or anti-ADM antibody fragment is
initiated and/or continued when the level of Pro-adrenomedullin or fragments
thereof
in said sample is above a certain threshold and/or wherein a treatment with an
anti-
ADM antibody or anti-ADM antibody fragment is withheld and/ or terminated if
the
said determined level of Pro-adrenomedullin or fragments thereof is below said

predetermined threshold.
17. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiment 14, wherein
treatment with an anti-ADM antibody or anti-ADM antibody fragment and/or
Angiotensin-Receptor-Agonists and/ or precursors thereof and/or inhibitors of
the
DPP3 activity is initiated and/or continued if the level of Pro-adrenomedullin
or
fragments thereof in said sample is above a certain threshold and said
determined level
of DPP3 is above said predetermined threshold of DPP3.
18. Vasopressor for use in therapy of shock in a subject that either runs into
shock or that
has developed shock, wherein said subject has a level of DPP3 in a sample of
bodily
fluid of said subject that is below a predetermined threshold, when determined
by a
method according to any of embodiments 1 ¨ 17.
19. Vasopressor for use in therapy of shock in a subject that either runs into
shock or that
has developed shock according to embodiment 18, wherein said vasopressor is
CA 03148275 2022-2-16

WO 2021/038078 59
PCT/EP2020/074134
selected from the group comprising dopamine, norepinephrine, a norepinephrine
equivalent, epinephrine, phenylephrine and vasopressin.
20. Vasopressor for use in therapy of shock in a subject that either runs into
shock or that
has developed shock according to embodiment 18 or 19, wherein said vasopressor
is
administered to said subject in a pharmaceutical formulation.
21. Vasopressor for use in therapy of shock in a subject that either runs into
shock or that
has developed shock according to any of embodiments 18 to 20, wherein said
subject
lo has a blood pressure of equal or less than 65mm Hg
22. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock, wherein said subject has a level
of DPP3
in a sample of bodily fluid of said subject that is above a predetermined
threshold
when determined by a method according to any of embodiments 1 ¨ 17.
23. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock according to embodiment 22,
wherein the
inhibitor of the activity of DPP3 is selected from the group comprising anti-
DPP3
antibody or anti-DPP3 antibody fragment or anti-DPP3 non-Ig scaffold.
24. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock according to embodiments 22 and
23,
wherein said inhibitor has a minimum binding affinity to DPP3 of equal or less
than
10-7 M.
25. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock according to any of embodiments 22
to
24, wherein the inhibitor of the activity of DPP3 is an antibody.
26. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock according to any of embodiments 22
to
25, wherein said inhibitor is an antibody that is monoclonal.
CA 03148275 2022-2-16

WO 2021/038078 60
PCT/EP2020/074134
27. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock according to any of embodiments 22
to
26, wherein said inhibitor is a monoclonal antibody, wherein the
complementarity
determining regions (CDR's) in the heavy chain comprises the sequences: SEQ ID
NO.: 7, SEQ ID NO.: 8 and/ or SEQ ID NO.: 9 and the complementarily
determining
regions (CDR's) in the light chain comprises the sequences: SEQ ID NO.: 10,
KVS
and/or SEQ ID NO.: 11.
28. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
lo
runs into shock or that has developed
shock according to any of embodiments 22 to
27, wherein said inhibitor is a humanized monoclonal antibody or humanized
monoclonal antibody fragment, wherein the heavy chain comprises the sequence:
SEQ
ID NO.: 12 and wherein the light chain comprises the sequence: SEQ ID NO.: 13.
29. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock according to any of embodiments 22
to
28, wherein said inhibitor is administered in combination with an Angiotensin-
Receptor-Agonist and/or precursor thereof.
30. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock according to embodiment 29,
wherein said
Angiotensin-Receptor-Agonist and/ or precursor thereof is selected from the
group
comprising angiotensin I, angiotensin II, angiotensin III, angiotensin IV, in
particular
angiotensin H.
31. Angiotensin-Receptor-Agonists and/ or precursors thereof and/or inhibitors
of the
DPP3 activity for use in the treatment of shock in a subject that either runs
into shock
or that has developed shock, wherein the treatment with said Angiotensin-
Receptor-
Agonists and/ or precursors thereof and/or inhibitors of the DPP3 activity is
initiated
and/or continued when the level of DPP3 in a sample of said subject is above a
certain
threshold and/or wherein a treatment with vasopressors is withheld and/ or
terminated
if said determined level of DPP3 is above said predetermined threshold.
CA 03148275 2022-2-16

WO 2021/038078 61
PCT/EP2020/074134
32. Vasopressor for use in the treatment of shock in a subject that either
runs into shock or
that has developed shock, wherein the treatment with said vasopressor is
initiated
and/or continued when the level of DPP3 in a sample of bodily fluid of said
subject is
below a certain threshold and/or wherein a treatment with Angiotensin-Receptor-

Agonists and/ or precursors thereof and/or inhibitors of the DPP3 activity is
withheld
and/ or terminated if the said determined level of DPP3 is below said
predetermined
threshold.
33. Vasopressor for use in the treatment of shock in a subject that either
runs into shock or
io
that has developed shock according to
embodiment 32, and wherein in addition the
level of Pro-adrenomedullin or fragments thereof is determined and wherein
treatment
with said anti-ADM antibody or anti-ADM antibody fragment is initiated and/or
continued when the level of Pro-adrenomedullin or fragments thereof in said
sample is
above a certain threshold and/or wherein a treatment with an anti-ADM antibody
or
anti-ADM antibody fragment is withheld an& or terminated if the said
determined
level of Pro-adrenomedullin or fragments thereof is below said predetermined
threshold.
34. Angiotensin-Receptor-Agonists and/ or precursors thereof and/or inhibitors
of the
DPP3 activity for use in the treatment of shock in a subject that either runs
into shock
or that has developed shock according to embodiment 31, and wherein in
addition the
level of Pro-adrenomedullin or fragments thereof is determined and wherein
treatment
with said anti-ADM antibody or anti-ADM antibody fragment is initiated and/or
continued when the level of Pro-adrenomedullin or fragments thereof in said
sample is
above a certain threshold and/or wherein a treatment with an anti-ADM antibody
or
anti-ADM antibody fragment is withheld and/ or terminated if the said
determined
level of Pro-adrenomedullin or fragments thereof is below said predetermined
threshold.
35, Anti-ADM antibody or anti-ADM antibody fragment for use in the treatment
of shock
in a subject that either runs into shock or that has developed shock according
to
embodiments 33-34, wherein treatment with said anti-ADM antibody or anti-ADM
antibody and/or Angiotensin-Receptor-Agonists and/ or precursors thereof
and/or
inhibitors of the DPP3 activity is initiated and/or continued if the level of
Pro-
CA 03148275 2022-2-16

WO 2021/038078 62
PCT/EP2020/074134
adrenomedullin or fragments thereof in said sample is above a certain
threshold and
said determined level of DPP3 is above said predetermined threshold of DPP3.
36. A method of treatment of shock in a subject that either runs into shock or
that has
developed shock, the method comprising administering vasopressor to said
subject,
wherein said subject has a level of DPP3 in a sample of bodily fluid of said
subject
that is below a predetermined threshold, when determined by a method according
to
any of embodiments 1 ¨ 17.
lo 37. A method of treatment of shock in a subject that either runs into
shock or that has
developed shock according to embodiment 35, the method comprising
administering
vasopressor to said subject,
wherein said vasopressor is selected from the group comprising dopamine,
norepinephrine, a norepinephrine equivalent, epinephrine, phenylephrine and
vasopressin.
38. A method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 35-36, the method comprising
administering vasopressor to said subject, wherein said vasopressor is
administered to
said subject in a pharmaceutical formulation.
39. A method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 35-37, the method comprising
administering vasopressor to said subject,
wherein said subject has a blood pressure of equal or less than 65mm Hg.
40. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock, the method comprising administering inhibitor of DPP3
activity to
said subject, wherein said subject has a level of DPP3 in a sample of bodily
fluid of
said subject that is above a predetermined threshold when determined by a
method
according to any of embodiments 1 ¨ 17.
41. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiment 39, the method comprising
administering
CA 03148275 2022-2-16

WO 2021/038078 63
PCT/EP2020/074134
an inhibitor of DPP3 activity to said subject, wherein the inhibitor of the
activity of
DPP3 is selected from the group comprising anti-DPP3 antibody or anti-DPP3
antibody fragment or anti-DPP3 non-Ig scaffold.
42. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 39-40, the method comprising
administering inhibitor of DPP3 activity to said subject, wherein said
inhibitor has a
minimum binding affinity to DPP3 of equal or less than 10-7 M.
lo
43. Method of treatment of shock in a
subject that either runs into shock or that has
developed shock according to embodiments 39-41, the method comprising
administering inhibitor of DPP3 activity to said subject, wherein the
inhibitor of the
activity of DPP3 is an antibody.
44. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 39-42, the method comprising
administering inhibitor of DPP3 activity to said subject, wherein said
inhibitor is an
antibody that is monoclonal.
45. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 39-43, the method comprising
administering inhibitor of DPP3 activity to said subject, wherein said
inhibitor is a
monoclonal antibody, wherein the complementarity determining regions (CDR's)
in
the heavy chain comprises the sequences: SEQ ID NO.: 7, SEQ ID NO.: 8 and/ or
SEQ ID NO.. 9 and the complementarity determining regions (CDR's) in the light
chain comprises the sequences: SEQ ID NO.: 10, KVS and/or SEQ lEs NO.: 11.
46. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 39-44, the method comprising
administering inhibitor of DPP3 activity to said subject, wherein said
inhibitor is a
humanized monoclonal antibody or humanized monoclonal antibody fragment,
wherein the heavy chain comprises the sequence: SEQ ID NO.: 12 and wherein the

light chain comprises the sequence: SEQ ID NO.: 13.
CA 03148275 2022-2-16

WO 2021/038078 64
PCT/EP2020/074134
47. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 39-45, the method comprising
administering an inhibitor of DPP3 activity to said subject, wherein said
inhibitor is
administered in combination with an Angiotensin-Receptor-Agonist and/ or
precursors
thereof.
48. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 39-46, the method comprising
lo
administering inhibitor of DPP3
activity to said subject, wherein said Angiotensin-
Receptor-Agonist and/ or precursors thereof is selected from the group
comprising
angiotensin I, angiotensin II, angiotensin III, angiotensin IV, in particular
angiotensin
49. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 39-47, the method comprising
administering Angiotensin-Receptor-Agonists and/ or precursors thereof and/or
inhibitor of DPP3 activity to said subject, wherein the treatment with said
Angiotensin-Receptor-Agonists and/or inhibitors of the DPP3 activity is
initiated
and/or continued when the level of DPP3 in a sample of said subject is above a
certain
threshold and/or wherein a treatment with vasopressors is withheld and/ or
terminated
if said determined level of DPP3 is above said predetermined threshold.
50. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 35-38, the method comprising
administering vasopressor to said subject, wherein the treatment with said
vasopressors is initiated and/or continued when the level of DPP3 in a sample
of
bodily fluid of said subject is below a certain threshold and/or wherein a
treatment
with Angiotensin-Receptor-Agonists and/ or precursors thereof and/or
inhibitors of
the DPP3 activity is withheld and/ or terminated if the said determined level
of DPP3
is below said predetermined threshold.
51. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiment 49, the method comprising
administering
CA 03148275 2022-2-16

WO 2021/038078 65
PCT/EP2020/074134
Angiotensin-Receptor-Agonists and/ or precursors thereof and/or inhibitor of
DPP3
activity to said subject, and wherein in addition the level of Pro-
adrenomedullin or
fragments thereof is determined and wherein treatment with said anti-ADM
antibody
or anti-ADM antibody fragment is initiated and/or continued when the level of
Pro-
s
adrenomedullin or fragments thereof in
said sample is above a certain threshold and/or
wherein a treatment with an anti-ADM antibody or anti-ADM antibody fragment is

withheld and/ or terminated if the said determined level of Pro-adrenomedullin
or
fragments thereof is below said predetermined threshold.
lo
52. Method of treatment of shock in a
subject that either runs into shock or that has
developed shock according to embodiment SO, the method comprising
administering
vasopressor to said subject, and wherein in addition the level of Pro-
adrenomedullin
or fragments thereof is determined and wherein treatment with said anti-ADM
antibody or anti-ADM antibody fragment is initiated and/or continued when the
level
15
of Pro-adrenomedullin or fragments
thereof in said sample is above a certain threshold
and/or wherein a treatment with an anti-ADM antibody or anti-ADM antibody
fragment is withheld and/ or terminated if the said determined level of Pro-
adrenomedullin or fragments thereof is below said predetermined threshold.
20
53. Method of treatment of shock in a
subject that either runs into shock or that has
developed shock according to embodiment 51 and 52, the method comprising
administering anti-ADM antibody or anti-ADM antibody fragment to said subject,

wherein in addition the level of Pro-adrenomedullin or fragments thereof is
determined and wherein treatment with said anti-ADM antibody or anti-ADM
25
antibody fragment is initiated and/or
continued when the level of Pro-adrenomedullin
or fragments thereof in said sample is above a certain threshold and/or
wherein a
treatment with an anti-ADM antibody or anti-ADM antibody fragment is withheld
and/ or terminated if the said determined level of Pro-adrenomedullin or
fragments
thereof is below said predetermined threshold
54. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 51 to 53, the method comprising
administering anti-ADM antibody or anti-ADM antibody fragment to said subject,

wherein treatment with said anti-ADM antibody or anti-ADM antibody and/or
CA 03148275 2022-2-16

WO 2021/038078 66
PCT/EP2020/074134
Angiotensin-Receptor-Agonists and/ or precursors thereof and/or inhibitors of
the
DPP3 activity is initiated and/or continued if the level of Pro-adrenomedullin
or
fragments thereof in said sample is above a certain threshold and said
determined level
of DPP3 is above said predetermined threshold of DPP3.
With the above context, the following consecutively numbered embodiments
provide further
specific aspects of the invention:
1. A method for predicting or diagnosing a refractory shock in a subject that
either runs
to into shock or that has developed shock, wherein said
method is comprising the steps:
= determining the level of DPP3 in a sample of bodily fluid of said
subject;
= comparing said level of determined DPP3 to a predetermined threshold,
wherein said subject is predicted to run into refractory shock or is diagnosed
as
having refractory shock if said determined level of DPP3 is above said
predetermined threshold.
2. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodimentl, wherein said
shock
is selected from the group comprising shock due to hypovolemia, cardiogenic
shock,
obstructive shock and distributive shock, in particular cardiogenic shock or
septic
shock.
3. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiment 1 and 2,
wherein
= in case of cardiogenic shock said subject may have suffered an acute
coronary
syndrome (e.g. acute myocardial infarction) or wherein said subject has heart
failure
(e.g. acute decompensated heart failure), myocarditis, arrhythmia,
cardiomyopathy,
valvular heart disease, aortic dissection with acute aortic stenos's,
traumatic chordal
rupture or massive pulmonary embolism or
= in case of hypovolemic shock said subject may have suffered a hemorrhagic
disease
including gastrointestinal bleed, trauma, vascular etiologies (e.g. ruptured
abdominal
aortic aneurysm, tumor eroding into a major blood vessel) and spontaneous
bleeding
CA 03148275 2022-2-16

WO 2021/038078 67
PCT/EP2020/074134
in the setting of anticoagulant use or a non-hemorrhagic disease including
vomiting,
diarrhea, renal loss, skin losses/insensible losses (e.g. bums, heat stroke)
or third-space
loss in the setting of pancreatitis, cirrhosis, intestinal obstruction,
trauma. or
= in case of obstructive shock said patient may have suffered a cardiac
tamponade,
tension pneumothorax, pulmonary embolism or aortic stenosis, or
= in case of distributive shock said patient may have septic shock,
neurogenic shock,
anaphylactic shock or shock due to adrenal crisis.
4. A method for predicting or diagnosing a refractory shock in a subject that
either runs
io into shock or that has developed shock according to embodiments 1
to 3, wherein said
method is used for initiation and/or termination and/ or stratification and/or
guidance
of treatment.
5. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to 4,
wherein a treatment is initiated and/or maintained and/or withheld and/ or
terminated
if said determined level of DPP3 is above said predetermined threshold.
6. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiment 5, wherein said
treatment is selected from the group of vasopressors, Angiotensin-Receptor-
Agonists
and/or precursors thereof, inhibitors of the DPP3 activity and anti-
adrenomedullin
antibodies or anti-adrenomedullin antibody fragments.
7. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to 6,

wherein either the level of DPP3 protein and/or the level of active DPP3 is
determined
and compared to a predetermined threshold, wherein the level of DPP3 is
determined
by contacting said sample of bodily fluid with a capture binder that binds
specifically
to DPP3.
8. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to any of embodiments 1 to 7,

wherein a treatment with Angiotensin-Receptor-Agonists and/ or precursors
thereof
CA 03148275 2022-2-16

WO 2021/038078 68
PCT/EP2020/074134
and/or inhibitors of the DPP3 activity is initiated and/or continued when the
level of
DPP3 in said sample is above a certain threshold and/or wherein a treatment
with
vasopressors is withheld and/ or terminated if said determined level of DPP3
is above
said predetermined threshold.
9. A method for predicting or diagnosing a refractory shock in a subject
that either runs
into shock or that has developed shock according to any of embodiments 1 to 7,

wherein a treatment with vasopressors is initiated and/or continued when the
level of
DPP3 in said sample is below a certain threshold and/or wherein a treatment
with
lo Angiotensin-Receptor-Agonists and/ or precursors
thereof and/or inhibitors of the
DPP3 activity is withheld and/ or terminated if the said determined level of
DPP3 is
below said predetermined threshold.
10. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiment 8, wherein in
addition the level of Pro-adrenomedullin or fragments thereof is determined
and
wherein treatment with an anti-ADM antibody or anti-ADM antibody fragment is
initiated and/or continued when the level of Pro-adrenomedullin or fragments
thereof
in said sample is above a certain threshold and/or wherein a treatment with an
anti-
ADM antibody or anti-ADM antibody fragment is withheld and/ or terminated if
the
said determined level of Pro-adrenomedullin or fragments thereof is below said

predetermined threshold.
11. A method for predicting or diagnosing a refractory shock in a subject that
either runs
into shock or that has developed shock according to embodiment 8, wherein
treatment
with an anti-ADM antibody or anti-ADM antibody fragment and/or Angiotensin-
Receptor-Agonists and/ or precursors thereof and/or inhibitors of the DPP3
activity is
initiated and/or continued if the level of Pro-adrenomedullin or fragments
thereof in
said sample is above a certain threshold and said determined level of DPP3 is
above
said predetermined threshold of DPP3.
12. Vasopressor for use in therapy of shock in a subject that either runs into
shock or that
has developed shock, wherein said subject has a level of DPP3 in a sample of
bodily
CA 03148275 2022-2-16

WO 2021/038078 69
PCT/EP2020/074134
fluid of said subject that is below a predetermined threshold, when determined
by a
method according to any of embodiments 1 ¨ 10.
13. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock, wherein said subject has a level
of DPP3
in a sample of bodily fluid of said subject that is above a predetermined
threshold
when determined by a method according to any of embodiments 1 ¨ 10, wherein
the
inhibitor of the activity of DPP3 is selected from the group comprising anti-
DPP3
antibody or anti-DPP3 antibody fragment or anti-DPP3 non-Ig scaffold.
Io
14. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock according to embodiment13, wherein
said
inhibitor is administered in combination with an Angiotensin-Receptor-Agonist
and/or
precursor thereof.
15. Inhibitor of the activity of DPP3 for use in therapy of shock in a subject
that either
runs into shock or that has developed shock according to embodiment 14,
wherein said
Angiotensin-Receptor-Agonist and/ or precursor thereof is selected from the
group
comprising angiotensin I, angiotensin It, angiotensin III, angiotensin IV, in
particular
angiotensin H.
16. A method of treatment of shock in a subject that either runs into shock or
that has
developed shock, the method comprising administering vasopressor to said
subject,
wherein said subject has a level of DPP3 in a sample of bodily fluid of said
subject
that is below a predetermined threshold, when determined by a method according
to
any of embodiments 1 ¨ 10.
17. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock, the method comprising administering inhibitor of DPP3
activity to
said subject, wherein said subject has a level of DPP3 in a sample of bodily
fluid of
said subject that is above a predetermined threshold when determined by a
method
according to any of embodiments 1 ¨ 10.
CA 03148275 2022-2-16

WO 2021/038078 70
PCT/EP2020/074134
18. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiment 17, the method comprising
administering
an inhibitor of DPP3 activity to said subject, wherein said inhibitor is
administered in
combination with an Angiotensin-Receptor-Agonist and/ or a precursor thereof.
19. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 16-17, the method comprising
administering Angiotensin-Receptor-Agonists and/ or precursors thereof and/or
inhibitor of DPP3 activity to said subject, wherein the treatment with said
lo Angiotensin-Receptor-Agonists and/or inhibitors of the DPP3 activity
is initiated
and/or continued when the level of DPP3 in a sample of said subject is above a
certain
threshold and/or wherein a treatment with vasopressors is withheld and/ or
terminated
if said determined level of DPP3 is above said predetermined threshold.
20. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiments 16-17, the method comprising
administering vasopressor to said subject, wherein the treatment with said
vasopressors is initiated and/or continued when the level of DPP3 in a sample
of
bodily fluid of said subject is below a certain threshold and/or wherein a
treatment
with Angiotensin-Receptor-Agonists and/ or precursors thereof and/or
inhibitors of
the DPP3 activity is withheld and/ or terminated if the said determined level
of DPP3
is below said predetermined threshold.
21. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiment 18, the method comprising
administering
Angiotensin-Receptor-Agonists and/ or precursors thereof ancUor inhibitor of
DPP3
activity to said subject, and wherein in addition the level of Pro-
adrenomedullin or
fragments thereof is determined and wherein treatment with said anti-ADM
antibody
or anti-ADM antibody fragment is initiated and/or continued when the level of
Pro-
adrenomedullin or fragments thereof in said sample is above a certain
threshold and/or
wherein a treatment with an anti-ADM antibody or anti-ADM antibody fragment is

withheld and/ or terminated if the said determined level of Pro-adrenomedullin
or
fragments thereof is below said predetermined threshold.
CA 03148275 2022-2-16

WO 2021/038078 71
PCT/EP2020/074134
22. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiment 19, the method comprising
administering
vasopressor to said subject, and wherein in addition the level of Pro-
adrenomedullin
or fragments thereof is determined and wherein treatment with said anti-ADM
antibody or anti-ADM antibody fragment is initiated and/or continued when the
level
of Pro-adrenomedullin or fragments thereof in said sample is above a certain
threshold
and/or wherein a treatment with an anti-ADM antibody or anti-ADM antibody
fragment is withheld and/ or terminated if the said determined level of Pro-
adrenomedullin or fragments thereof is below said predetermined threshold.
ID
23. Method of treatment of shock in a subject that either runs into shock or
that has
developed shock according to embodiment 20 and 21, the method comprising
administering anti-ADM antibody or anti-ADM antibody fragment to said subject,

wherein in addition the level of Pro-adrenomedullin or fragments thereof is
determined and wherein treatment with said anti-ADM antibody or anti-ADM
antibody fragment is initiated and/or continued when the level of Pro-
adrenomedullin
or fragments thereof in said sample is above a certain threshold and/or
wherein a
treatment with an anti-ADM antibody or anti-ADM antibody fragment is withheld
and/ or terminated if the said determined level of Proeadrenomedullin or
fragments
thereof is below said predetermined threshold.
24. A method for prognosing an outcome and/or the risk of an adverse event in
a subject
that has developed refractory shock, wherein said method is comprising the
steps:
= determining the level of DPP3 in a sample of bodily fluid of said subject;
= comparing said level of determined DPP3 to a predetermined threshold,
= correlating said level of DPP3 with said risk of an adverse event in said

subject, wherein an elevated level above a certain threshold is predictive for
an
enhanced risk of said adverse events or,
= correlating said level of DPP3 with success of a therapy or intervention in
said
subject, wherein a level below a certain threshold is predictive for a success
of
therapy or intervention.
CA 03148275 2022-2-16

WO 2021/038078 72
PCT/EP2020/074134
EXAMPLES
Example 1 ¨ Methods for the measurement of DPP 3 protein and DPP3 activity
Generation of antibodies and determination DPP3 binding ability: Several
murine antibodies
were produced and screened by their ability of binding human DPP3 in a
specific binding
assay (see Table 1).
Peptides/ conjugates for immunization:
DPP3 peptides for immunization were synthesized, see Table 1, (JPT
Technologies, Berlin,
io Germany) with an additional N-terminal cystein (if no cystein is present
within the selected
DPP3-sequence) residue for conjugation of the peptides to Bovine Serum Albumin
(BSA).
The peptides were covalently linked to BSA by using Sulfolink-coupling gel
(Perbio-science,
Bonn, Germany). The coupling procedure was performed according to the manual
of Perbio.
Recombinant GST-hDPP3 was produced by USBio (United States Biological, Salem,
MA,
USA).
Immunization of mice, immune cell fusion and screening:
Balb/c mice were intraperitoneally (i.p.) injected with 84 pg GST-hDPP3 or 100
pg
DPP3-peptide-BSA-conjugates at day 0 (emulsified in TiterMax Gold Adjuvant),
84 pg or
100 pg at day 14 (emulsified in complete Freund's adjuvant) and 42 pg or 50
lig at day 21
and 28 (in incomplete Freund's adjuvant). At day 49 the animal received an
intravenous (i.v.)
injection of 42 pg GST-hDPP3 or 50 pg DPP3-peptide-BSA-conjugates dissolved in
saline.
Three days later the mice were sacrificed and the immune cell fusion was
performed.
Splenocytes from the immunized mice and cells of the myeloma cell line SP2/0
were fused
with 1 ml 50% polyethylene glycol for 30 s at 37 C. After washing, the cells
were seeded in
96-well cell culture plates. Hybrid clones were selected by growing in HAT
medium [RPMI
1640 culture medium supplemented with 20% fetal calf serum and HAT-
Supplement]. After
one week, the HAT medium was replaced with HT Medium for three passages
followed by
returning to the normal cell culture medium.
The cell culture supernatants were primarily screened for recombinant DPP3
binding IgG
antibodies two weeks after fusion. Therefore, recombinant GST-tagged hDPP3
(USBiologicals, Salem, USA) was immobilized in 96-well plates (100 ng/ well)
and incubated
with 50 I cell culture supernatant per well for 2 hours at room temperature.
After washing of
CA 03148275 2022-2-16

WO 2021/038078 73
PCT/EP2020/074134
the plate, 50 jtl / well POD-rabbit anti mouse IgG was added and incubated for
1 h at RT
After a next washing step, 50 pl of a chromogen solution (3,7 m.M o-
phenylendiamine in
citrate/ hydrogen phosphate buffer, 0.012% 11202) were added to each well,
incubated for 15
minutes at RT and the chromogenic reaction stopped by the addition of 50 pl 4N
sulfuric acid.
Absorption was detected at 490 mm.
The positive tested microcultures were transferred into 24-well plates for
propagation. After
retesting the selected cultures were cloned and re-cloned using the limiting-
dilution technique
and the isotypes were determined.
Mouse monoclonal antibody production
io Antibodies raised against GST-tagged human DPP3 or DPP3-peptides were
produced via
standard antibody production methods (Marx et aL 1997) and purified via
Protein A. The
antibody purities were? 90% based on SDS gel electrophoresis analysis.
Characterization of antibodies ¨ binding to hDPP3 and/ or immunization peptide

To analyze the capability of DPP3/ immunization peptide binding by the
different antibodies
and antibody clones a binding assay was performed:
a) Solid phase
Recombinant GST-taggedhDPP3 (SEQ ID NO. 1) or a DPP3 peptide (immunization
peptide,
SEQ ID NO. 2) was immobilized onto a high binding microliter plate surface (96-
Well
polystyrene microplates, Greiner Bio-One international AG, Austria, 1 pg/well
in coupling
buffer [50 mM Tris, 100 niM NaCl, pH7,8], lb at RT). After blocking with 5%
bovine serum
albumin, the microplates were vacuum dried.
Li) Labelling procedure (Tracer)
100 mg (100 itl) of the different antiDPP3 antibodies (detection antibody, 1
mg/ ml in PBS,
pH 7.4) were mixed with 10 pl acridinium NHS-ester (1 mg/ml in acetonitrile,
InVent GmbH,
Germany; EP 0 353 971) and incubated for 30 min at room temperature. Labelled
antiDPP3
antibody was purified by gel-filtration HPLC on Shodex Protein 5 pm KW-803
(Showa
Denko, Japan). The purified labeled antibody was diluted in assay buffer (50
mmo1/1
potassium phosphate, 100 mmo1/1 NaC1, 10 mmo1/1 Na2-EDTA, 5 g/1 bovine serum
albumin,
1 Wl murine IgG, 1 8/1 bovine IgG, 50 grnol/1 amastatin, 100 p.mo1/1
leupeptin, pH 7.4). The
final concentration was approx. 5-7*106 relative light units (RUT) of labelled
compound
CA 03148275 2022-2-16

WO 2021/038078 74
PCT/EP2020/074134
(approx. 20 ng labeled antibody) per 200 1. acridinium ester
chemiluminescence was
measured by using a Centro LB 960 luminometer (Berthold Technologies GmbH &
Co. KG).
c) hDPP3 binding assay
The plates were filled with 200 pl of labeled and diluted detection antibody
(tracer) and
incubated for 2-4 h at 2-8 'C. Unbound tracer was removed by washing 4 times
with 350 pl
washing solution (20 inM PBS, pH 7.4, 0.1 % Triton X-100). Well-bound
chemiluminescence
was measured by using the Centro LB 960 luminometer (Berthold Technologies
GmbH & Co
KG).
Characterization of antibodies ¨ hDPP3-inhibition analysis
io To analyze the capability of DPP3 inhibition by the different antibodies
and antibody clones a
DPP3 activity assay with known procedure (Jones et at., 1982) was performed
Recombinant
GST-tagged hDPP3 was diluted in assay buffer (25 ng/ ml GST-DPP3 in 50 m114
Tris-HCI,
pH7,5 and 100 pM ZnC12) and 200 gl of this solution incubated with 10 pig of
the respective
antibody at room temperature. After 1 hour of pre-incubation, fluorogenic
substrate Arg-Arg-
f3NA (20 gl, 2m1v1) was added to the solution and the generation of free DNA
over time was
monitored using the Twinkle LB 970 microplate fluorometer (Berthold
Technologies GmbH
& Co. KG) at 37 C. Fluorescence of fiNA is detected by exciting at 340 nm and
measuring
emission at 410 nm. Slopes (in RFU/ min) of increasing fluorescence of the
different samples
are calculated. The slope of GST-hDPP3 with buffer control is appointed as 100
% activity.
The inhibitory ability of a possible capture-binder is defined as the decrease
of GST-hDPP3
activity by incubation with said capture-binder in percent.
The following table represents a selection of obtained antibodies and their
binding rate in
Relative Light Units (RLU) as well as their relative inhibitory ability (%;
table 1). The
monoclonal antibodies raised against the below depicted DPP3 regions, were
selected by their
ability to bind recombinant DPP3 and/ or immunization peptide, as well as by
their inhibitory
potential.
All antibodies raised against the GST-tagged, full length form of recombinant
hDPP3 show a
strong binding to immobilized GST-taggedhDPP3. Also antibodies raised against
the SEQ ID
NO.: 2 peptide bind to GST-hDPP3. The SEQ ID NO.: 2 antibodies also strongly
bind to the
immunization peptide.
CA 03148275 2022-2-16

WO 2021/038078 75
PCT/EP2020/074134
2552 3.053.621 0
65%
SEQ ID GST tagged recombinant FL-
2553 3.777.985 0 35%1_737
NO.: 1 hDPP3
2554 1.733.815 0 30%
2555 3.805363 0
25%
1963 141.822 2.163.038 60%
1964 100.802 2.041.928 60%
1965 99.493 1.986394 70%
SEQ ID 474-
CETV1NPETGEQIQSWYRSGE
1966 118.097 1. 990. 702 65%
NO.: 2 493
1967 113.736 1.909.954 70%
1968 105.696 2.017.731 65%
1969 82.558 2.224.025 70%
Table 1: list of antibodies raised against full-length or sequences of hDPP3
and their ability to
bind hDPP3 (SEQ ID NO.: 1) or immunization peptide (SEQ ID NO.: 2) in RLU, as
well as
the maximum inhibition of recombinant GST-hDPP3.
The development of a luminescence immunoassay for the quantification of DPP3
protein
concentrations (DPP3-LIA) as well as an enzyme capture activity assay for the
quantification
of DPP3 activity (DPP3-ECA) have been described recently (Rehfeld et al. 2018.
JAW in
press), which is incorporated here in its entirety by reference.
io Example 2 - DPP3 for prognosis of short-term mortality
DPP3 concentration in plasma of patients with sepsis/ septic shock and
cardiogenic shock was
determined and related to the short term-mortality of the patients.
Study Cohort ¨ Sepsis/Septic Shock
In 574 plasma samples from patients of the Adrenomedullin and Outcome in
Severe Sepsis
and Septic Shock (AdrenOSS) study DPP3 was measured. AdrenOSS is a
prospective,
observational, multinational study including 583 patients admitted to the
intensive care unit
CA 03148275 2022-2-16

WO 2021/038078 76
PCT/EP2020/074134
with sepsis or septic shock (Hollinger et al., 2018). 292 patients were
diagnosed with septic
shock.
Study Cohort ¨ Cardiogenic Shock
Plasma samples from 108 patients that were diagnosed with cardiogenic shock
were screened
for DPP3. Blood was drawn within 6 h from detection of cardiogenic shock.
Mortality was
followed for 7 days.
hDPP3 immunoassay:
An immunoassay (LIA) or an activity assays (ECA) detecting the amount of human
DPP3
io (LIA) or the activity of human DPP3 (ECA), respectively, was used for
determining the DPP3
level in patient plasma. Antibody immobilization, labelling and incubation
were performed as
described in Rehfeld et al. (Rehfeld et al. 2018).
Results
Short-term patients' survival in sepsis patients was related to the DPP3
plasma concentration
at admission. Patients with DPP3 plasma concentration above 40.5 ng/mL (3rd
Quartile) had
an increased mortality risk compared to patients with DPP3 plasma
concentrations below this
threshold (Figure 1A). Applying this cut-off to the subcohort of septic shock
patients,
revealed an even more pronounced risk for short-torm mortality in relation to
high DPP3
plasma concentrations (Figure 1B). When the same cut-off is applied to
patients with
cardiogenic shock, also an increased risk for short-term mortality within 7
days is observed in
patients with high DPP3 (Figure IC).
Example 3 - Purification of human native DPP3
Human erythrocyte lysate was applied on a total of 100 ml of Sepahrose 4B
resin (Sigma-
Aldrich) and the flow through was collected. The resin was washed with a total
of 370 mL
PBS buffer, pH 7.4 and the wash fraction was combined with the collected flow
through,
resulting in a total volume of 2370 mL.
For the immuno-affinity purification step, 110 mg of monoclonal anti-hDPP3 mAb
AK2552
were coupled to 25.5 mL of UltraLink Hydrazide Resin (Thermo Fisher
Scientific) according
to the manufacturer's protocol (GlycoLink Immobilization Kit, Thermo Fisher
Scientific).
The coupling efficiency was 98%, determined by quantification of uncoupled
antibody via
CA 03148275 2022-2-16

WO 2021/038078 77
PCT/EP2020/074134
Bradford-technique. The resin-antibody conjugate was equilibrated with 10 bed
volumes of
wash-binding buffer (PBS, 0.1% TritonX-100, pH 7.4), combined with 2370 mL of
cleared
red blood cell lysate and incubated at 4 C under continuous stirring for 2h.
Consequently,
100 mL of the incubation mixture was spread on ten 15 mL polypropylene columns
and the
flow-through was collected by centrifugation at 1000xg for 30 seconds. This
step was
repeated several times resulting in 2.5 mL of DPP3-loaded resin per column.
Each column
was washed 5 times with 10 mL of wash-binding buffer using the gravity-glow
approach.
DPP3 was eluted by placing each column in 15-mL falcon tube containing 2 mL of

neutralization buffer (1M Tris-HCI, pH 8.0), followed by addition of 10 mL of
elution buffer
to (100 rtiM Glycine-HO, 0.1% TritonX-100, pH 3.5) per column and
immediate centrifugation
for 30 seconds at 1000xg. The elution step was repeated 3 times in total
resulting in 360 mL
of combined eluates. The pH of the neutralized eluates was 8Ø
The combined eluates were loaded on a 5 mL HiTrap Q-sephare HP column (GE
Healthcare)
equilibrated with lEX-buffer Al (100 m1VI Glycine, 150 InM Tris, pH 8.0) using
the sample
IS pump of the Aida Start system (GE Healthcare). After sample
loading, the column was
washed with five column volumes of 1:EX Buffer A2 (12 mIVI NaH2Pa4, PH 7.4) to
remove
unbound protein. Elution of DPP3 was achieved by applying a sodium chloride
gradient over
column volumes (50 mL) in a range of 0 ¨ 1 M NaCl using IEX-buffer B (12 mM
NaH2PO4,1 M NaCl, pH 7.4). The eluates were collected in 2 mL fractions.
Buffers used for
ion exchange chromatography were sterile filtered using a 0.22 [tM bottle-top
filter.
A purification table with the respective yields and activities of each
purification step is given
in table 2. Figure 2 shows an SDS-PAGE on a gradient gel (4-20%) of native
hDPP3 purified
from human erythrocyte lysate.
Table 2: Purification of DPP 3 from human erythrocytes
Step DPP3 Total Total
Yield in Specific Purification
amount in protein in activity in %
activity in factor
% (LIA) mgt) p moUmin
U/mge)
(ECA)c)
Lysate 100 204160 55
100 0.00027 -
IAP 80.6 71.2 46.1
84 0.65 2407
IEX 75 6.6 38.7
70 5.9 21852
a) Relative DPP3 amount was determined in all fractions using the DPP3-LIA
assay. Amount of DPP3
in starting material was set to 100% and remaining DPP3 amount in purification
fractions was
correlated to the starting material.
CA 03148275 2022-2-16

WO 2021/038078 78
PCT/EP2020/074134
b) Total protein amount was determined using the method of Lowry modified by
Peterson (Peterson
1977. Analytical Biochemistry 356:346-356).
`) Total Arg2-BNA hydrolyzing activity in limol of substrate converted per
minute was determined
using the DPP3-ECA, calibrated via B-naphtylamine (0,05-100 M).
d) Purification yield was calculated form total Arg2-BNA hydrolyzing activity.
Arg2-BNA hydrolyzing
activity in starting material was set to 100%.
e) Specific activity is defined as limo' of substrate converted per minute and
mg of total protein.
I) The purification factor is the quotient of specific activities after and
before each purification step.
io Example 4 - Effect of native DP1'3 in an animal model
The effect of native hDPP3 injection in healthy mice was studied by monitoring
the
shortening fraction and renal resistive index.
Wild type Black 6 mice (8-12 weeks, group size refer to table 3) were
acclimated during 2
is weeks and a baseline echocardiography was done. The mice were randomly
allocated to one
of the two groups and, subsequently, native DPP3 protein or PBS were injected
intravenously
via a retro-orbital injection with a dose of 600 ftg/kg for DPP3 protein.
After DPP3 or PBS injection, cardiac function was assessed by echocardiography
(Gao et al.
20 2011) and renal function assessed by renal resistive index (Lubas el
al., 2014. Dewitte et al,
2012) at 15, 60 and 120 minutes (Figure 3).
Group Number of Animals Treatment
WT PBS 3 PBS
WT+DPP3 4 Native
DPP3
Table 3: list of experiment groups
Results
The mice treated with native DPP3 protein show significantly reduced
shortening fraction
25 compared to the the control group injected with PBS (Fig 4A). The
WT+DPP3 group also
displays worsening renal function as observed by the renal resistive index
increase (Figure
4B).
CA 03148275 2022-2-16

WO 2021/038078 79
PCT/EP2020/074134
Example 5¨ Development of Procizumab
Antibodies raised against SEQ ID NO. 2 were characterized in more detail
(epitope mapping,
binding affinities, specificity, inhibitory potential). Here the results for
clone 1967 of SEQ ID
NO.: 2 (AK1967; "Procizumab") are shown as an example.
Determination of AK1967 epitope on DPP3:
For epitope mapping of AK1967 a number of N- or C-terminally biotinylated
peptides were
synthesized (PE GmbH, Hennigsdorf, Germany). These peptides include the
sequence of the
full immunization peptide (SEQ ID No. 2) or fragments thereof, with stepwise
removal of one
io amino acid from either C- or N-terminus (see table 5 for a complete list
of peptides).
High binding 96 well plates were coated with 2 pg Avidin per well (Greiner Die-
One
international AG, Austria) in coupling buffer (500 tuM Tris-HCl, pH 72, 100
inM NaCI).
Afterwards plates were washed and filled with specific solutions of
biotinylated peptides
(10 ng/ well; buffer ¨ 1xPBS with 0.5% BSA). Anti-DPP3 antibody AK1967 was
labelled
with a chemiluminescence label according to Example 1.
The plates were filled with 200 1 of labeled and diluted detection antibody
(tracer) and
incubated for 4 h at room temperature. Unbound tracer was removed by washing 4
times with
350 I washing solution (20 mM PBS, pH 7.4, 0.1 % Triton X-100). Well-bound
chemiluminescence was measured by using the Centro LB 960 luminometer
(Berthold
Technologies GmbH & Co. KG). Binding of AK1967 to the respective peptides is
determined
by evaluation of the relative light units (RLU). Any peptide that shows a
significantly higher
RLU signal than the unspecific binding of AK1967 is defined as AK1967 binder.
The
combinatorial analysis of binding and non-binding peptides reveals the
specific DPP3 epitope
of AK1967.
Determination of binding affinities using Octet:
The experiment was performed using Octet Red96 (ForteBio). AK1967 was captured
on
kinetic grade anti-humanFc (AHC) biosensors. The loaded biosensors were then
dipped into a
dilution series of recombinant GST-tagged human DPP3 (100, 33.3, 11.1, 3.7
TIME
Association was observed for 120 seconds followed by 180 seconds of
dissociation. The
buffers used for the experiment are depicted in table 4. Kinetic analysis was
performed using
a 1:1 binding model and global fitting.
CA 03148275 2022-2-16

WO 2021/038078 80
PCT/EP2020/074134
Buffer Composition
Assay Buffer PBS with 0.1% BSA,
0.02% Tween-21
Regeneration Buffer 10 rnIVI Glycine buffer
(pH 1.7)
Neutralization Buffer PBS with 0.1% BSA,
0.02% Tween-21
Table 4: Buffers used for Octet measurements
Western Blot analysis of Binding specificity of AK1967:
Blood cells from human EDTA-blood were washed (3x in PBS), diluted in PBS and
lysed by
repeated freeze-thaw-cycles. The blood cell lysate had a total protein
concentration of
250 lig/mt, and a DPP3 concentration of 10 pig/ml. Dilutions of blood cell
lysate (1:40, 1:80,
1:160 and 1:320) and of purified recombinant human His-DPP3 (31.25-500 nem')
were
subjected to SDS-PAGE and Western Blot. The blots were incubated in 1.)
blocking buffer
(bcPBS-T with 5% skim milk powder), 2.) primary antibody solution (AK1967
1:2.000 in
io blocking buffer) and 3.) IMP labelled secondary antibody (goat anti
mouse IgG, 1:1.000 in
blocking buffer). Bound secondary antibody was detected using the Amersham ECL
Western
Blotting Detection Reagent and the Amersham Imager 600 UV (both from GE
Healthcare).
DPP3 inhibition assay:
To analyze the capability of DPP3 inhibition by AK1967 a DPP3 activity assay
with known
is procedure clones et al., 1982) was performed as described in example 1. The
inhibitory
ability AK1967 is defined as the decrease of GST-hDPP3 activity by incubation
with said
antibody in percent. The resulting lowered DPP3 activities are shown in an
inhibition curve in
Figure 1C.
Epitope mapping:
20 The analysis of peptides that AK1967 binds to and does not bind to
revealed the DPP3
sequence INPETG (SEQ ID No.: 3) as necessary epitope for AK1967 binding (see
table 5).
peptide
AK1967
peptide sequence
binding
#1 bio afnfdqetvinpetgeqiqsw y r s
g yes
#2 bio afnfdqetvinpetgeqiq
yes
#3 bio afnfdqetvinpetgeqi
yes
#4 bio afnfdqetvinpetgeq
yes
CA 03148275 2022-2-16

WO 2021/038078 81
PCT/EP2020/074134
#5 bio afnfdqetvinpetge
yes
#6 bio afnfdqetvinpetg
yes
#7 bio afnfdqetvinpet
no
#8 bio afnfdqetvinpe
no
#9 bio afnfdqetvinp
no
#10 bio afnfdqetv in
no
#11
etgeqiqsw y kbio no
#12
petgeqiqsw y kbio .. 110
#13
npetgeqiqsw ykbio no
#14
inpetgeqiqsw y kbio yes
#15
vinpetgeqiqsw y kbio yes
#16
tvinpetgeqiqsw y kbio yes
#17
etvinpetgeqiqsw y kbio yes
Table 5: Peptides used for Epitope mapping of AK1967
Binding affinity:
AK1967 binds with an affinity of 2.24(104 M to recombinant GST-hDPP3 (kinetic
curves see
Figure 5).
Specificity and inhibitory potential:
The only protein detected with AK1967 as primary antibody in lysate of blood
cells was
DPP3 at 80 kDa (Figure 6) The total protein concentration of the lysate was
250 pg/m1
whereas the estimated DPP3 concentration is about 10 pg/ml. Even though there
is 25 times
io more unspecific protein in the lysate, AK1967 binds and detects
specifically DPP3 and no
other unspecific binding takes place.
AK1967 inhibits 15 ng/ ml DPP3 in a specific DPP3 activity assay with an IC50
of about
rig/ml (Figure 7).
Chi merization/ Humanization:
IS The monoclonal antibody AK1967 ("Procizumab"), with the ability of
inhibiting DPP3
activity by 70 %, was chosen as possible therapeutic antibody and was also
used as template
for chimerization and humanization.
CA 03148275 2022-2-16

WO 2021/038078 82
PCT/EP2020/074134
Humanization of murine antibodies may be conducted according to the following
procedure:
For humanization of an antibody of murine origin the antibody sequence is
analyzed for the
structural interaction of framework regions (FR) with the complementary
determining regions
(CDR) and the antigen. Based on structural modelling an appropriate FR of
human origin is
selected and the murine CDR sequences are transplanted into the human FR.
Variations in the
amino acid sequence of the CDRs or FRs may be introduced to regain structural
interactions,
which were abolished by the species switch for the FR sequences. This recovery
of structural
interactions may be achieved by random approach using phage display libraries
or via
directed approach guided by molecular modeling (Altnagro and Fransson, 2008.
Humanization of antibodies. Front Biosci. 13:1619-33).
With the above context, the variable region can be connected to any subclass
of constant
regions (IgG, IgM, IgE. IgA), or only scaffolds, Fab fragments, Fv, Fab and
F(ab)2. In
example 6 and 7 below, the murine antibody variant with an IgG2a backbone was
used. For
chimerization and humanization a human IgGlx backbone was used.
IS For epitope binding, only the CDRs are of importance. The CDRs for the
heavy chain and the
light chain of the murine anti-DPP3 antibody (AK1967) are shown in SEQ ID No.
6, SEQ ID
No. 7 and SEQ ID No. 8 for the heavy chain and SEQ II) No. 9, sequence KVS and
SEQ
No. 10 for the light chain, respectively. Sequencing of the anti-DPP3 antibody
(AK1967)
revealed an antibody heavy chain variable region (H chain) according to SEQ ID
No.: 11 and
an antibody light chain variable region (L chain) according to SEQ ID No.: 12.
Example 6¨ Effect of Procizumab in septic shock-induced heart failure
In this experiment, the effect of Procizumab injection in sepsis-induced heart
failure rats
(Rittirsch et al. 2009) was studied by monitoring the shortening fraction
Cecal ligation puncture (CLP) model of septic shock:
Male Wistar rats (2-3 months, 300 to 400 g, group size refers to table 6) from
the Centre
d'elevage Janvier (France) were allocated randomly to one of three groups. All
animals were
anesthetized using ketamine hydrochloride (90 mg/ kg) and xylazine (9 mg/ kg)
intra-
peritoneally (ill). For induction of polymicrobial sepsis, CLP was performed
using
Rittirsch's protocol with minor modifications. A ventral midline incision (1.5
cm) was made
to allow exteriorization of the cecum. The cecum is then ligated just below
the ileocecal valve
CA 03148275 2022-2-16

WO 2021/038078 83
PCT/EP2020/074134
and punctured once with an 18-gauge needle. The abdominal cavity is then
closed in two
layers, followed by fluid resuscitation (3 ml/ 100 g body of weight of saline
injected
subcutaneously) and returning the animal to its cage. Sham animals were
subjected to surgery,
without getting their cecum punctured. CLP animals were randomized between
placebo and
therapeutic antibody.
Study design:
The study flow is depicted in Figure 8. After CLP or sham surgery the animals
were allowed
to rest for 20 hours with free access to water and food. Afterwards they were
anesthetized,
tracheotomy done and arterial and venous line laid. At 24 hours after CLP
surgery either
io AK1967 or vehicle (saline) were administered with 5 mg/kg as a bolus
injection followed by
a 3h infusion with 7.5 mg/kg. As a safety measure, hemodynamics were monitored
invasively
and continuously from t =0 till 3 h.
At t=O (baseline) all CLP animals are in septic shock and developed a decrease
in heart
function (low blood pressure, low shortening fraction). At this time point
Procizumab or
vehicle (PBS) were injected (i.v.) and saline infusion was started. There were
1 control group
and 2 CLP groups which are summarized in the table below (table 6). At the end
of the
experiment, the animals were euthanized, and organs harvested for subsequent
analysis.
Group Number of Animals CLP
Treatment
Sham 7 No
PBS
CLP-PBS 6 Yes
PBS
CLP-PCZ 4 Yes
PCZ
Table 6: list of experimental groups
Invasive Blood Pressure:
Hemodynamic variables were obtained using the AcqKnowledge system (BIOPAC
Systems,
Inc., USA). It provides a fully automated blood pressure analysis system. The
catheter is
connected to the BIOPAC system through a pressure sensor.
For the procedure, rats were anesthetized (ketamine and xylazine). Animals
were moved to
the heating pad for the desired body temperature to 37-37.5 C. The
temperature feedback
probe was inserted into the rectum. The rats were placed on the operating
table in a supine
position. The trachea was opened and a catheter (16G) was inserted for an
external ventilator
CA 03148275 2022-2-16

WO 2021/038078 84
PCT/EP2020/074134
without to damage carotid arteries and vagus nerves. The arterial catheter was
inserted into
the right carotid artery. The carotid artery is separate from vagus before
ligation. A central
venous catheter was inserted through the left jugular vein allowing
administration of PCZ or
PBS.
Following surgery, the animals were allowed to rest for the stable condition
prior to
hemodynamic measurements. Then baseline blood pressure (BP) were recorded.
During the
data collection, saline infusion via arterial line was stopped.
Echocardiography:
Animals were anesthetized using ketamine hydrochloride. Chests were shaved and
rats were
io placed in decubitus position. For transthoracic echocardiographic (TTE)
examination a
commercial GE Healthcare Vivid 7 Ultra-sound System equipped with a high
frequency (14-
MHz) linear probe and 10-MHz cardiac probe was used. All examinations were
recorded
digitally and stored for subsequent off-line analysis.
Grey scale images were recorded at a depth of 2 cm. Two-dimensional
examinations were
initiated in a parasternal long axis view to measure the aortic annulus
diameter and the
pulmonary artery diameter. M-mode was also employed to measure left
ventricular (LV)
dimensions and assess fractional shortening (FS%). LVFS was calculated as LV
end-diastolic
diameter - LV end-systolic diameter / LV end-diastolic diameter and expressed
in %. The
time of end-diastole was therefore defined at the maximal diameter of the LV.
Accordingly,
end-systole was defined as the minimal diameter in the same heart cycle. All
parameters were
measured manually. Three heart cycles were averaged for each measurement.
From the same parasternal long axis view, pulmonary artery flow was recorded
using pulsed
wave Doppler. Velocity time integral of pulmonary artery outflow was measured.
From an apical five-chamber view, mitral flow was recorded using pulsed
Doppler at the level
of the tip of the mitral valves.
Results:
The septic shock-induced heart failure rats treated with PBS (CLP+PBS) show
reduced
shortening fraction compared to the sham animals (Fig. 9A). The CLP+PBS group
also
displays high mortality rate (Fig. 9B). In contrast, application of Procizumab
to sep-induced
heart failure rats improves shortening fraction (Fig. 9A) and drastically
reduces the mortality
rate (Fig. 9B).
CA 03148275 2022-2-16

WO 2021/038078 85
PCT/EP2020/074134
Example 7 - Effect of Procizumab on heart and kidney function
The effect of Procizumab in isoproterenol-induced heart failure in mice was
studied by
monitoring the shortening fraction and renal resistive index.
Isoproterenol-induced cardiac stress in mice:
Acute heart failure was induced in male mice at 3 months of age by two daily
subcutaneous
injections of 300 mg/kg of Isoproterenol, a non-selective 13-adrenergic
agonist
(DL-Isoproterenol hydrochloride, Sigma Chemical Co) (ISO) for two days (Verger
et al,
2016). ISO dilution was performed in NaCl 0.9%. ISO-treated mice were randomly
assigned
io to two groups (Table 7) and PBS or Procizumab (10 mg/kg) were injected
intravenously after
baseline echocardiography (Gao et at. 2011) and renal resistive index
measurements (Lubas
et al. 2014. Dew/tie etal. 2012) were performed at day 3 (Figure 10 A and B).
Cardiac function was assessed by echocardiography (Gao et al., 2011) and by
the renal
resistive index (Lubas et at, 2014. Dew/tie et al, 2012) at 1 hour, 6 hours
and 24 hours
is (Figure 10 A and B). The group of mice that was injected with vehicle
(PBS) instead of
isoproterenol was subjected to no further pharmacological treatment and served
as the control
group (Table 7).
Group Number of Animals Treatment
Sham+PBS 27 PBS
HF+PBS 15 PBS
HF+PCZ 20
Procizumab (PCZ)
Table 7: list of experimental groups
Results:
Application of Procizumab to isoproterenol-induced heart failure mice restores
heart function
within the first hour after administration (Fig. 11A). Kidney function of sick
mice shows
significant improvement at 6 hours post Procizumab injection and is comparable
to the kidney
function of sham animals at 24 hours (Fig. 11B).
CA 03148275 2022-2-16

WO 2021/038078 86
PCT/EP2020/074134
Example 8 - DPP3 indicates vasopressor need and response to vasopressor
therapy
DPP3 concentrations in plasma of septic shock patients were determined using a
hDPP3
immunoassay and related to the need for vasopressor therapy.
Study Cohort ¨ Septic Shock
Plasma samples from 292 patients that were diagnosed with septic shock from
the AdrenOSS
(see example 2) study were screened for DPP3, Human DPP3 was measured as
described in
Example 1,
0 Results
Patients with an increased need to vasopressor administration for more than 5
days had high
plasma concentrations of DPP3 (Figure 12A). In contrast, patients that
responded to
vasopressor administration and where vasopressor administration could be
discontinued
within the first 5 days, had significantly reduced plasma DPP3 concentrations
(Figure 12B),
This indicates that development of refractory shock and thus an increased need
of vasopressor
administration due to a diminished response of this therapy, is related to
high DPP3 plasma
concentrations in patients with septic shock.
Patients with vasopressor-resistant refractory septic shock (noradrenaline
>0.5 mg/kg/min)
show significantly higher plasma DPP3 concentrations than compared to patients
that
required noradrenaline doses of < 0.5 lig/kg/min (p<0.001) (Figure 16A),
Moreover, the
DPP3 plasma concentration was strongly associated with mortality in patients
with
vasopressor-resistant refractory septic shock (Figure 16B).
Example 9¨ DPP3 is related to refractory shock
DPP3 concentrations in plasma of cardiogenic shock patients were determined
using a hDPP3
immunoassay and related to the development of refractory shock.
Study Cohort ¨ Cardiogenic Shock
Plasma samples from 57 patients that were diagnosed with cardiogenic shock
after acute
myocardial infarction were screened for DPP3. Human DPP3 was measured as
described in
Example 1.
Results
CA 03148275 2022-2-16

WO 2021/038078 87
PCT/EP2020/074134
Patients with a DPP3 plasma concentration above a certain threshold on
admission (59.1
ng/ml; 3rd quartile) developed refractory shock to a higher extent (47%) than
patients with
DPP3 plasma concentrations below 59.1 ng/ml (12%) (Figure 13).
Example 10¨ DPP3 and bio-ADM indicate short-term mortality in shock
DPP3 and bio-ADM concentrations in plasma of septic shock patients were
determined using
a hDPP3 and a bio-ADM immunoassay and related to the short term-mortality of
the patients.
io Study Cohort ¨ Septic Shock
Plasma samples from 292 patients that were diagnosed with septic shock from
the AdrenOSS
study were screened for DPP3 and bio-ADM. Human DPP3 was measured as described
in
Example 1. Bio-ADM was measured as decribed in Weber et al. (Weber et al.
2017. JADVI
2(2): 1-4).
Results
Plasma concentrations of bio-ADM and DPP3 were measured in septic schock
patients.
Patients were grouped according to specific cut-offs determined to be the 3rd
quartile of all
measured plasma concentrations of the respective marker (Table 8). Equal
numbers of
patients had either high DPP3 only or high bio-ADM only (15.4%), while a
lesser extent
showed elevated plasma concentrations in both, bio-ADM and DPP3 (9.6%).
DPP3 low (<48.4 ng/ml) DPP3 high (> 48.4 ng/ml)
bio-ADM low (< 213 pg/mL) 174 (59.6%)
45 (15.4%)
bio-ADM high (>213 pg/mL) 45 (15.4%)
28 (9.6%)
Table 8: Patient numbers with low/high DPP3 and low/high bio-ADM. Cut-offs
were
assigned based on the Q3 (highest 25%) for both.
Mortality within the first 4 weeks after admission was related to the bio-ADM
and DPP3
concentration on admission. Patients with either only a high bio-ADM or only a
high DPP3
plasma concentration had a substantial increased risk to die within the first
4 weeks compared
to patients that had a plasma concentration of either bio-ADM (Figure 14A) or
either DPP3
CA 03148275 2022-2-16

WO 2021/038078 88
PCT/EP2020/074134
(Figure 14B) below a certain threshold (3"I quartile). Survival in patients
with high bio-ADM
was better than survival in patients with high DPP3.
When both markers, bio-ADM and DPP3, were combined, an even higher risk for
short-term
mortality was identified in comparison to the mortality that was related to an
increase of one
of both markers only (Figure 14C). Merely 28.6% of the patients with high bio-
ADM and
high-DPP3 plasma concentrations survived the first 4 weeks after admission.
Example 11 ¨ DPP3 and bio-ADM DPP3 indicates vasopressor need and response to

vasopressor therapy
DPP3 and bio-ADM concentrations in plasma of septic shock patients were
determined using
a hDPP3 and a bio-ADM immunoassay and related to the need for vasopressor
therapy.
Study Cohort ¨ Septic Shock
In 292 plasma samples from patients that were diagnosed with septic shock from
the
is AdrenOSS study DPP3 and bio-ADM concentrations were measured. Human DPP3
was
measured as described in Example 1. Bio-ADM was measured as decribed in Weber
et al.
(Weber et al. 2017. JALA, 120): 1-4).
Results
Plasma concentrations of bio-ADM and DPP3 were measured in septic schock
patients.
Patients were grouped according to specific cut-offs determined as the 3rd
quartile of all
measured plasma concentrations of the respective marker in the septic shock
cohort (low
DPP3 <48.4 ng/mL, low bio-ADM <213 pg/mL; high bio-ADM 213 pg/mL; high DPP3
48.4 ng/mL).
Patients with high DPP3, but low bio-ADM plasma concentrations had a higher
need for
consecutive vasopressor administration compared to patients with either low
DPP3 + low bio-
ADM or low DPP3 + high bio-ADM (Figure 15; Table 9). In contrast, patients
that had low
DPP3 and low bio-ADM plasma concentrations, or patients that had low DPP3, but
high bio-
ADM plasma concentrations on admission could be discontinued of vasopressor
administration earlier than patients with high DPP3 plasma concentrations on
admission
(Figure 15; Table 9). This indicates that vasopressor therapy in septic shock
patients with high
bio-ADM can be discontinued earlier due to a better therapeutic response
compared to septic
CA 03148275 2022-2-16

WO 2021/038078 89
PCT/EP2020/074134
shock patients with high DPP3. These patients (high DPP3) need longer
treatment with
vasopressors due to non-responsiveness.
Days with - pn low
DPP3 high DPP3 nigh
vasopressor need DPP3 law
,
bIoADM low mbloADNI high bioADM1ow b
1 42 8
5 2
2 49 11
6 2
3 25 6
8 2
4 16 4
1 2
8 3 2 0
6 3 1
6 1
7 31 12
17 19
Table 9: Patients with vasopressor need grouped according to their DPP3 and
bio-ADM
plasma concentration at admission. Cut-offs were assigned based on the Q3
(highest 25% of
5 determined values) for both; low DPP3 <48.4 ng/mL, low bio-ADM
<213 pg/mL; high bio-
ADM > 213 pg/mL; high DPP3 48.4 ng/mL; 7: ?7 days on vasopressor or dead
within 7
days
CA 03148275 2022-2-16

WO 2021/038078 90
PCT/EP2020/074134
EXAMPLES OF SEQUENCES
SEQ lID No. 1 ¨ hDPP3 aa 1-737
MADTQYILPNDIGVSSLDCREAFRLL SPTERLYAYHL SRAAWYGGLAVLLQTSPEAP
YIYALLSRLFRAQDPDQLRQHALAEGLTEEEYQAFLVYAAGVYSNMGNYK SFGDTK
FVPNLPKEKLERVILGSEAAQQHPEEVRGLWQTCGELMFSLEPRLRHLGLGKEGITTY
F SGNCTMEDAKLAQDFLD SQNL SA YNTRLFKEVDGEGKPYYEVRL AS VLGSEPSLD S
EVTSKLKSYEFRGSPFQVTRGDYAPILQKVVEQLEKAKAYAANSHQGQMLAQY1ESF
TQGSIEAHKSGSRFWIQDKGPIVESYIGFIESYRDPFGSRGEFEGFVAVVNKAMSAKFE
lo RLVASAEQLLICELPWPPTFEKDICFLTPDFTSLDVLTFAGSGIPAGINIPNYDDLRQTEG
FKNVSLGNVLAVAYATQREKLTFLEEDDICDLYILWKGP SFDVQVGLHELLGHGSGK
LFVQDEKGAFNFDQETWNPETGEQIQ SW YRSGETWD SKF STIASSYEECRAESVGLY
LCLHPQVLEIFGFEGADAEDVIYVNWLNMVRAGLLALEFYTPEAFNWRQAHMQARF
VILRVLLEAGEGLVTITPTTGSDGRPDARVRLDRSICIRSVGKPALERFLRRLQVLKSTG
IS DVAGGRALYEGYATVTDAPPECFLTLRDTVLLRICE SRKLIVQPNTRLEGSDVQLLEY
EASAAGLIRSFSERFPEDGPELEFILTQLATADARFWKGPSEAPSGQA
SEQ ID No. 2 ¨ hDPP3 an 474-493 (N-Cys) ¨ immunization peptide with additional
N-
terminal Cystein
20 CETVINPETGEQIQSWYRSGE
SEQ ID No. 3¨ hDPP3 aa 477-482 ¨ epitope of AK1967
INPETG
25 SEQ ID No. 4¨ variable region of murine AK1967 in heavy chain
QVTLICESGPGILQP SQ TL SLTC SF SGF SLSTSGMSVGWIRQPSGKGLEWLAHIWWNDN
KSYNPALK SRLTISRDTSNNQVFLKIASVVT ADTGTYFCARNYSYDYWGQGTTLTVS
30 SEQ ID No. 5¨ variable region of in urine AK1967 in light chain
DVVVTQTPL SLSVSLGDPASI SCRS SRSLVHSIGSTYLHWYL QKPGQSPKLLIYKV SNR
FSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYFC SQSTHVPWTFGGGTICLEIK
CA 03148275 2022-2-16

WO 2021/038078 91
PCT/EP2020/074134
SEQ ID No. 6¨ CDR1 of murine AK1967 in heavy chain
GFSLSTSGMS
SEQ ID No. 7¨ CDR2 of murine AK1967 in heavy chain
IWWNDNK
SEQ ID No. 8¨ CDR 3 of murine AK1967 in heavy chain
ARNYSYDY
1 o SEQ ID No. 9¨ CDR1 of murine AK1967 in light chain
RSLVHSIGSTY
CDR2 of murine AK1967 in light chain
KVS
SEQ ID No. 10- CDR3 of murine AK1967 in light chain
SQSTHVPWT
SEQ ID No. 11 ¨ humanized AK1967 ¨ heavy chain sequence (IgGIK backbone)
MDPKGSLSWRILLFLSLAFELSYGQITLKESGPTLVICPTQTLTLTCTFSGFSLSTSGMS
VGWIRQPPGKALEWLABIWWNDNKSYNPALKSRLTITRDTSKNQVVLTMTNMDPV
DTGTYYCARNYSYDYWGQGTLVTVS S ASTKGP SVFPLAPS SK ST SGGTAALGCLVK
DYFPEPVTVSWNSGALTSGVHTFPAVLQ SSGLYSLS SVVTVP SS SLGTQTYICNVNHK
PSNTKVDICKVEPK SCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVV
VDVSHEDPEVICFNWYVDGVEVHNAKTICPREEQYNSTYRVVSVLTVLHQDWLNGICE
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVICGFYPSDI
AVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVN1HEALH
NHYTQKSLSLSPG
SEQ ID No. 12¨ humanized AK1967 ¨ light chain sequence (IgGht backbone)
METDTLLLWVLLLWVPGSTGDIVNITQTPLSLSVTPGQPASISCKSSRSLVHSIGSTYLY
WYLQKPGQSPQLLIYKVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCSQST
HVPWTFGGGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWK
CA 03148275 2022-2-16

WO 2021/038078 92
PCT/EP2020/074134
VDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHICVYACEVTHQGLSSPVT
KSFNRGEC
SEQ ID No. 13¨ Angiotensin if (synonyme: 5-isoleucine-angiotensin II)
DRVYIHPF
SEQ ID No. 14¨ Angiotensin 11 analogue (5-valine-angiotensin II)
DRVYVH:PF
io SEQ ID No. 15¨ Angiotensin if analogue (Asnl-Phe4)
NRVFIHPF
SEQ ID No. 16¨ Angiotensin II hexapeptide
VYIHPF
SEQ ID No. 17¨ Angiotensin It nonapeptide
NRVYYVHPF
SEQ ID No. 18¨ Angiotensin II analogue (Asnl-fies-Deshingiotensin II)
NRVYIHPI
SEQ ID No. 19¨ Angiotensin 11 analogue (tAsni-Des-Alal-angiotensin 11)
NRVYIH:PA
SEQ ID No. 20¨ Angiotensin if analogue (1Asni-diiodoTyr4-De5kangiotensin II
NRVYIHPF
SEQ ID No. 21 ¨ Angiotensin III
RVYIHPF
SEQ ID No. 22¨ Angiotensin HI analogue (Valtangiotensin III)
RVYVHPF
CA 03148275 2022-2-16

WO 2021/038078 93
PCT/EP2020/074134
SEQ ID No. 23- Angiotensin III analogue (Phe3-angietensin III)
RVFIHPF
SEQ ID No. 24¨ Angiotensin HI analogue (Ille4-Ala71-angiotensin HI)
RVYMPA
SEQ ID No. 25¨ Angiotensin HI analogue (diiodoTyr3-11e1-angiotensin HI)
RVYMPF
io SEQ ID No. 26¨ Angiotensin IV
VYMPF
SEQ ID No. 27¨ Angiotensin IV analogue (Va13-angiotensin IV)
VYVHPF
SEQ ID No. 28¨ Angiotensin IV analogue (Phel-angiotensin IV)
VFIHPF
SEQ ID No. 29- Angiotensin IV analogue (Ille3-Ala6]-angiotensin IV)
VYIHPA
SEQ ID No. 30- Angiotensin IV analogue (diiodoTyr2-11e3-angiotensin IV)
VYMPF
SEQ ID No. 31 (proADM): 164 amino acids (22 ¨ 185 of preproADM)
ARLDVASEF RKKWNKWALS RGKRELRMSS SYPTGLADVK AGPAQTURP
QDMKGASRSP EDSSPDAARI RVKRYRQSMN NFQGLRSFGC RFGTCTVQICL
AHQIYQFTDK DKDNVAPRSK ISPQGYGRRR RRSLPEAGPG RTLVSSKPQA
HGAPAPPSGS APHFL
SEQ ID No. 32 (Proadrenomedullin N-20 terminal peptide, PAMP): amino acids 22
¨ 41
of preproADM
ARLDVASEF R1CKWNKWALS R
CA 03148275 2022-2-16

WO 2021/038078 94
PCT/EP2020/074134
SEQ ID No. 33 (Midregional proAdrenomedullin, MR-proADM): amino acids 45 ¨ 92
of
preproADM
ELRMSS SYPTGLADVK AGPAQTLIRP QDMKGASRSP EDSSPDAARI RV
SEQ ID No. 34 (mature Adrenomedullin (mature ADM); amidated ADM; bio-ADM;
hADM): amino acids 95¨ 146 -CONH2
YRQSMN NFQGLRSFGC RFGTCTVQKL AHQIYQFTDK DKDNVAPRSK ISPQGY -
CONH2
io SEQ ID No. 35 (Adrenomedullin 1-52-Gly (ADM 1-52-Gly)): amino
acids 95 ¨ 147 of
preproADM
YRQSMN NFQGLRSFGC RFGTCTVQICL AHQIYQFTDK DKDNVAPRSK ISPQGYG
SEQ ID No. 36 (C-terminal proAdrenomedullin, CT-proADM): amino acids 148 ¨ 185
is of preproADM
RRR RRSLPEAGPG RTLVSSKPQA HGAPAPPSGS APHFL
SEQ ID No. 37 (N-terminal part of mature ADM): amino acids 1-21 of mature ADM
YRQSMNNFQGLRSFGCRFGTC
SEQ ID No. 38 (CDR1 heavy chain anti-ADM antibody)
GYTFSRYW
SEQ ID No. 39 (CDR2 heavy chain anti-ADM antibody)
ILPGSGST
SEQ ID No. 40 (CDR3 heavy chain anti-ADM antibody)
TEGYEYDGFDY
SEQ ID No. 41 (CDR1 light chain anti-ADM antibody)
QSIVYSNGNTY
SEQ ID No. 42 (CDR3 light chain anti-ADM antibody)
FQGSHTPYT
CA 03148275 2022-2-16

WO 2021/038078 95
PCT/EP2020/074134
SEQ ID No. 43 (anti-ADM antibody (Adrecizumab) heavy chain)
QVQLVQSGAEVKICPGSSVKVSCKASGYTFSRYWIEWVRQAPGQGLEWIGELLPGSGS
TNYNQKFQGRVTITADTSTSTAYMELSSLRSEDTAVYYCTEGYEYDGFDYWGQGTT
VTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTF
PAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVFPKSCDKTHTCPP
CPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVICFNWYVDGVEVH
NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKG
QPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD
io SDGSFTLYSKLTVDKSRWQQGNVFSCSVMHEALIINHYTQKSLSLSPGK
SEQ ID NO: 44 (anti-ADM antibody (Adrecizumab) light chain)
DVVLTQSPLSLPVTLGQPASISCRSSOSIVYSNGNTYLEWYLQRPGQSPRLLIYRVSNR
FSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCFOGSHIPYTFGGGTKLEIKRTVAA
PSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSK
DSTYSLSSTLTLSKADYEICHKVYACEVTHQGLSSPVTKSFNRGEC
CA 03148275 2022-2-16

WO 2021/038078 96
PCT/EP2020/074134
FIGURE DESCRIPTION
Figure 1: Kaplan-Meier survival plots in relation to low (<40.5 ng/mL) and
high (> 40.5
ng/mL) DPP3 concentrations. (A) 7-Day survival of patients with sepsis in
relation to DPP3
plasma concentration; (B) 7-Day survival of Patients with cardiogenic shock in
relation to
DPP3 plasma concentration; (C) 7-Day survival of patients with septic shock in
relation to
DPP3 plasma concentration.
Figure 2: SDS-PAGE on a gradient gel (4-20%) of native hDPP3 purified from
human
io erythrocyte lysate. Molecular weight marker is indicated as arrows.
Figure 3: Experimental design - Effect of native DPP3 in an animal model.
Figure 4: (A) DPP3 injection causes shortening fraction reduction and
therefore leads to
deteriorating heart function. (B) Decreased kidney function is also observed
via increased
renal resistive index.
Figure 5: Association- and dissociation curve of the AK1967-DPP3 binding
analysis using
Octet. AK1967 loaded biosensors were dipped into a dilution series of
recombinant
GST-tagged human DPP3 (100, 33.3, 11.1, 3.7 n.M) and association and
dissociation
monitored.
Figure 6: Western Blot of dilutions of blood cell lysate and detection of DPP3
with AK1967
as primary antibody.
Figure 7: Inhibition curve of native DPP3 from blood cells with inhibitory
antibody AK1967.
Inhibition of DPP3 by a specific antibody is concentration dependent, with an
IC50 at
¨15 ng/ml when analyzed against 15 ng/ml DPP3.
Figure 8: Experimental setup ¨ Effect of Procizumab in sepsis-induced heart
failure.
Figure 9: Procizumab drastically improves shortening fraction (A) and
mortality rate (B) in
sepsis-induced heart failure rats.
CA 03148275 2022-2-16

WO 2021/038078 97
PCT/EP2020/074134
Figure 10: Experimental design - Isoproterenol-induced cardiac stress in mice
followed by
Procizumab treatment (B) and control (A).
Figure 11: Procizumab improved shortening fraction (A) and reduced the renal
resistive
index (B) within 1 hour and 6 hours after administration, respectively, in
isoproterenol-
induced heart failure mice.
Figure 12: (A) Days with need of vasopressor therapy in septic shock patients
in relation to
io DPP3 plasma concentrations >7 days on vasopressor or dead within
7 days; * p<0.05 in post
hoc comparison vs. groups 1-4 (B) Need of vasopressor therapy in septic shock
patients in
relation to DPP3 plasma concentrations. Patients with vasopressor therapy for
maximal 5 days
(<5), and patients with vasopressor therapy longer than 5 days or that died
within 7 days were
grouped together (>5).
Figure 13: DPP3 is related to refractory shock. High DPP3 concentrations in
plasma of
cardiogenic shock patients are related with a higher risk to develop
refractory cardiogenic
shock compared to patients that have DPP3 plasma concentrations below a
certain threshold.
Figure 14: Kaplan-Meier survival plots (A) 4-week survival of patients with
septic shock in
relation to bio-ADM plasma concentration; values are grouped in quartiles (B)
4-week
survival of patients with septic shock in relation to DPP3 plasma
concentration; values are
grouped in quartiles (C) 4-week survival of patients with septic shock in
relation to bio-ADM
and DPP3 plasma concentration; Cut-offs were determined based on 31d quartile
of all
measured plasma concentrations of the respective marker: both low DPP3 <48.4
ng/mL, bio-
ADM <213 pg/mL; bio-ADM high? 213 pg/mL; DPP3 high? 48.4 ng/mL.
Figure 15: Patients with vasopressor need grouped according to their DPP3 and
bio-ADM
plasma concentration on admission. Proportions of patients receiving
vasopressors for 1-7
days are shown in greyscale - lighter colors represent longer treatment
duration. Cut-offs were
assigned based on the Q3 (highest 25% of determined values) for both; low DPP3
<48.4
ng/mL, low bio-ADM < 213 pg/mL; high bio-ADM > 213 pg/mL; high DPP3 > 48.4
ng/mL;
7: >7 days on vasopressor or dead within 7 days.
CA 03148275 2022-2-16

WO 2021/038078 98
PCT/EP2020/074134
Figure 16: (A) Plasma DPP3 concentration in patients with septic refractory
shock who are in
need of the vasopressor noradrenalin below (n=186) and above (n=95) 0.5
pg/kg/min
(p<0.001). (B) Kaplan-Meier survival plots (A) 4-week survival of patients
with septic
refractory shock in relation to DP133 plasma concentration; values are grouped
in quartiles.
REFERENCES
1. Hollenberg et at. Ann Intern Med 1999; 131:47-59.
2. Reynolds HR, Hochman JS. Circulation 2008; 117:686-697.
Io
3. Tarvasmald et al. Eur Heart Fail John Wiley & Sons, Ltd; 2018; 20:572-
581.
4. Thiele Et at. NEng1.JMed 2012; 367:1287-1296.
5. Champion S. Eur J Heart Fail 2018; 20:197-198.
6. Hochman JS. Circulation 2003; 107:2998-3002.
7. Shah eta]. Clin its Cardiol 2018;107:287-303.
8. Schmidt etal. Eur Heart J 2015;36:2246¨ 2256.
9. Muller et at. Intensive Care Med 2016; 42:370-378.
10. Chen et at. Cr!: Care 2016; 20:336.
11. Harjola et at. Eur J Heart Fail John Wiley & Sons, Ltd; 2015; 17:501-
509.
12. Harjola et al. Eur J Heart Fail John Wiley & Sons, Ltd; 2018; 20:1081-
1099.
13. Bakker et al. Am J Surg 1996; 171:221-226.
14. Attalla et at. Acute Card Care 2012;14:20-26.
CA 03148275 2022-2-16

WO 2021/038078 99
PCT/EP2020/074134
15. Zhang Z, Xu X. Grit Care Med 2014; 42:2118-2125.
16. Allardet at. J Neurochem 1987; 48:1553-1559.
17. Prajapati SC, Chauhan SS. FEBS J 2011; 278:3256-3276.
18. Ocaranza MP, Jalil JE. Hypertens (Dallas, Tex 1979) 2016; 68:552-554.
19. Rehfeld L, J Appl Lab Med 2019 JAL111 3(6):943-953.
20. Deniau et al. Eur J Heart Fail 2020 Feb;22(2):290-299.
21. Levy J Am Coll Cardiol 2018; 72:173-182.
22. Kohsalca et al. Arch Intern Med 2005; 165:1643-1650.
23. Hochmanet at. N Engl J Med 1999; 341:625-634.
24. Thiele et at. N Engl J Med 2012; 367:1287-1296.
25. Thiele et al. N Engl J Med 2017; 377:2419-2432.
26. The TRIUMPH Investigators*, Alexander JH,et at. JAMA 2007; 297:1657.
27. Reyentovich et al. Nat Rev Cardiol 2016; 13:481-492.
28. Mebazaa et al. Intensive Care Med 2018; 44:760-773.
29. Bassi et at Grit Care Res Pract 2013; 2013:654708.
30. Baran et al. Catheter Cardiovasc Intery 2019; 94:29-37.
CA 03148275 2022-2-16

Representative Drawing

Sorry, the representative drawing for patent document number 3148275 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2020-08-28
(87) PCT Publication Date 2021-03-04
(85) National Entry 2022-02-16
Examination Requested 2022-09-08

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $100.00 was received on 2023-08-10


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2024-08-28 $50.00
Next Payment if standard fee 2024-08-28 $125.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $407.18 2022-02-16
Maintenance Fee - Application - New Act 2 2022-08-29 $100.00 2022-02-16
Request for Examination 2024-08-28 $814.37 2022-09-08
Maintenance Fee - Application - New Act 3 2023-08-28 $100.00 2023-08-10
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
4TEEN4 PHARMACEUTICALS GMBH
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
National Entry Request 2022-02-16 1 38
Miscellaneous correspondence 2022-02-16 8 367
Miscellaneous correspondence 2022-02-16 3 65
Miscellaneous correspondence 2022-02-16 11 539
Miscellaneous correspondence 2022-02-16 6 249
Drawings 2022-02-16 15 239
Priority Request - PCT 2022-02-16 141 5,722
Claims 2022-02-16 5 239
International Search Report 2022-02-16 3 81
Patent Cooperation Treaty (PCT) 2022-02-16 1 55
Description 2022-02-16 99 4,465
Priority Request - PCT 2022-02-16 143 5,798
Patent Cooperation Treaty (PCT) 2022-02-16 1 51
Correspondence 2022-02-16 2 46
Abstract 2022-02-16 1 16
National Entry Request 2022-02-16 9 176
Sequence Listing - New Application / Sequence Listing - Amendment 2022-02-16 4 104
Change to the Method of Correspondence 2022-02-16 4 104
Cover Page 2022-05-18 1 37
Request for Examination 2022-09-08 3 106
Amendment 2024-03-20 35 1,591
Description 2024-03-20 100 4,810
Claims 2024-03-20 6 366
Examiner Requisition 2023-11-21 4 250

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :